Abstract

Adenosine-to-inosine (A-to-I) RNA editing is a prevalent RNA modification essential for cell survival. The process is catalyzed by the adenosine deaminase acting on RNA (ADAR) enzyme family that converts adenosines in double-stranded RNAs (dsRNAs) into inosines, which are read as guanosines during translation. Deep sequencing has helped to reveal that A-to-I editing occurs across various types of RNAs, affecting their functions. RNA editing detection is now so sophisticated that we can achieve a high level of accuracy and sensitivity to identify low-abundance edited events. Consequently, A-to-I editing has been implicated in various biological processes, including immune and stress responses, cancer progression, and stem cell fate determination. In particular, a crucial role for this process has been recently reported in hematopoietic cell development and hematologic malignancy progression. Results from genetic mouse models have demonstrated the impact of ADARs' catalytic activity on hematopoietic cells, complemented by insights from human cell studies. Meanwhile, clinical studies have implicated ADAR enzymes and RNA editing events in hematologic malignancies and highlighted their potential as prognostic indicators. In this review, we outline the regulatory mechanisms of RNA editing in both normal hematopoiesis and hematologic malignancies. We then speculate on how targeting ADAR expression and site-specific RNA substrates might serve as a therapeutic avenue for affected patients.

To date, >170 RNA modifications have been identified across various RNA types, playing a crucial role in enhancing transcriptomic and proteomic diversity by regulating splicing, messenger RNA (mRNA) stability, codon decoding, and the functions of noncoding RNAs.1 RNA editing is one of the most prevalent posttranscriptional RNA modification that serves to introduce changes to RNA sequences.2 In vertebrates, RNA editing includes adenosine-to-inosine (A-to-I) editing, catalyzed by the ADAR family of enzymes, and cytidine-to-uracil (C-to-U), catalyzed by Apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC) family of enzymes.3 This review primarily focuses on A-to-I RNA editing, with a brief mention of C-to-U RNA editing.

The ADAR family comprises 3 members: ADAR1, ADAR2, and ADAR3.4 ADAR1 and ADAR2 are broadly expressed and share common functional domains, including 2 to 3 repeats of the double-stranded RNA (dsRNA)–binding domain and a catalytic deaminase domain.5-7 ADAR3 is specifically expressed in the brain and has no documented deaminase activity.8 ADAR1 has 2 isoforms: an interferon (IFN)–inducible p150 isoform and a constitutively expressed p110 isoform.9 ADAR1 p110 is predominantly localized in the nucleus; however, studies have shown that it can shuttle between the nucleus and cytoplasm under specific conditions, although less dynamically than ADAR1 p150.10 The dynamic nucleocytoplasmic shuttling of ADAR1p150 is facilitated by a nuclear export signal located within helix α1 of its Zα domain.11 

The primary function of ADAR enzymes is to regulate immune responses by editing host RNA to mark it as “self” while leaving viral RNA unedited as "nonself." Unedited dsRNAs are recognized by immune sensors such as melanoma differentiation-associated protein 5 (MDA5) and dsRNA-activated protein kinase R (PKR), which trigger signaling cascades that lead to the production of IFNs and IFN-stimulated genes (ISGs).12,13 ADARs can also facilitate immune evasion by introducing coding changes that promote tumor growth while concurrently shielding tumor cells by suppressing ISG-driven immune responses.14-16 Additionally, ADARs have been implicated in regulating stress response17 and stem cell fate.18,19 

Hematopoiesis is a finely regulated process orchestrated by lineage-specific transcription factors that direct the generation of mature cells from hematopoietic stem and progenitor cells (HSPCs). HSPC maintenance, self-renewal, differentiation, and lineage commitment are controlled by diverse posttranscriptional regulators.20,21 Posttranscriptional perturbations are, therefore, closely associated with the development of hematologic malignancies.22 A key example is seen in the transition from pre–leukemic stem cells (pre-LSCs) to LSCs.20,21,23 ADAR has been extensively studied in this context, being implicated in both normal and aberrant hematopoiesis.24,25 Among the ADAR family, ADAR1 is the most well established in its roles, whereas ADAR2, although less studied, is gradually emerging as a molecule of increasing interest in leukemia research. Despite mounting evidence, a narrative review of ADAR’s role within the hematopoietic system is lacking. Here, we provide an overview of RNA editing in normal and abnormal hematopoiesis. We focus on critical RNA editing sites and RNA editing detection methods. We then highlight the latest evidence that show the relevance of RNA editing in normal and pathological hematopoiesis. Finally, we speculate on how RNA editing might be leveraged as a potential therapeutic strategy.

To dissect the function and implications of A-to-I editing in hematopoiesis, it is important to first understand the underlying mechanism of this RNA editing event (Figure 1). ADARs preferentially edit mismatched adenosines within imperfect dsRNA, converting them to inosines.26,27 The inosine is then recognized as guanosine by the cellular translation and splicing machinery.28 This editing activity is particularly prominent in Alu repeats in humans, which constitute ∼11% of the genome. In contrast, the mouse genome is devoid of Alu repeats.29 

Figure 1.

The role of ADAR enzymes in RNA editing. ADAR enzymes bind to dsRNA regions and convert A to I, affecting various RNA types. (A) Precursor mRNA. Editing within exons can alter coding sequences, leading to amino acid changes or premature translation termination, potentially resulting in dysfunctional proteins. Editing at introns can influence splicing, generating extended or shortened isoforms. In the 3' UTR, editing affects miRNA binding, in which perfect binding with new target miRNAs promotes mRNA degradation and translation repression, whereas imperfect binding with previously bound miRNAs may enhance mRNA stability. (B) Mature mRNA. In some cases, RNA editing occurs after splicing, in which dsRNA structures within a single exon or between exons can lead to amino acid substitutions. (C) LncRNA. Editing of lncRNAs modulates their interactions with miRNAs and influences their stability by recruiting RNA-stabilizing proteins. (D) Pri-miRNA. Editing at (1) the Drosha-DGCR8 cleavage site or (2) the DICER/TRBP cleavage site can inhibit miRNA biogenesis, leading to degradation by inosine-dependent ribonucleases, such as Tudor-SN. Additionally, editing may hinder (3) RISC loading. Editing within (4) the seed region can alter miRNA target specificity, enabling binding to new target genes or releasing previous target genes. In the figure, the red strand represents the guide strand, whereas the blue strand represents the passenger strand. (E) Circular RNA. RNA editing in inverted Alu repeats of circular RNAs can unwind the dsRNA structure, preventing 3′ to 5′ back splicing of exons and thus promoting linear mRNA formation. (F) Viral RNA. RNA editing in viral RNA induces amino acid changes, playing a critical role in the viral life cycle, immune evasion, and pathogenesis. (G) tRNA. RNA editing in the anticodon loop (eg, A34 and A37) enables inosine to pair with multiple codons. circRNA, circular RNA; Drosha, mouse protein; DROSHA, human protein. lncRNA, long noncoding RNA; RISC, RNA-induced silencing complex; tRNA, transfer RNA.

Figure 1.

The role of ADAR enzymes in RNA editing. ADAR enzymes bind to dsRNA regions and convert A to I, affecting various RNA types. (A) Precursor mRNA. Editing within exons can alter coding sequences, leading to amino acid changes or premature translation termination, potentially resulting in dysfunctional proteins. Editing at introns can influence splicing, generating extended or shortened isoforms. In the 3' UTR, editing affects miRNA binding, in which perfect binding with new target miRNAs promotes mRNA degradation and translation repression, whereas imperfect binding with previously bound miRNAs may enhance mRNA stability. (B) Mature mRNA. In some cases, RNA editing occurs after splicing, in which dsRNA structures within a single exon or between exons can lead to amino acid substitutions. (C) LncRNA. Editing of lncRNAs modulates their interactions with miRNAs and influences their stability by recruiting RNA-stabilizing proteins. (D) Pri-miRNA. Editing at (1) the Drosha-DGCR8 cleavage site or (2) the DICER/TRBP cleavage site can inhibit miRNA biogenesis, leading to degradation by inosine-dependent ribonucleases, such as Tudor-SN. Additionally, editing may hinder (3) RISC loading. Editing within (4) the seed region can alter miRNA target specificity, enabling binding to new target genes or releasing previous target genes. In the figure, the red strand represents the guide strand, whereas the blue strand represents the passenger strand. (E) Circular RNA. RNA editing in inverted Alu repeats of circular RNAs can unwind the dsRNA structure, preventing 3′ to 5′ back splicing of exons and thus promoting linear mRNA formation. (F) Viral RNA. RNA editing in viral RNA induces amino acid changes, playing a critical role in the viral life cycle, immune evasion, and pathogenesis. (G) tRNA. RNA editing in the anticodon loop (eg, A34 and A37) enables inosine to pair with multiple codons. circRNA, circular RNA; Drosha, mouse protein; DROSHA, human protein. lncRNA, long noncoding RNA; RISC, RNA-induced silencing complex; tRNA, transfer RNA.

Close modal

The effects of A-to-I editing are impactful when it occurs in coding regions, because they can alter the amino acid sequence and modify protein function.24,30-32 RNA editing events occur more frequently, however, in noncoding regions that consist of inversely oriented repetitive elements. RNA editing in introns or in exon-intron junctions can lead to aberrant splicing, generating new isoforms.33-36 Additionally, A-to-I modification in the 3' untranslated region (3′ UTR) can affect mRNA translation or expression by influencing microRNA (miRNA) binding specificity.37,38 In some cases, RNA editing occurs after splicing (mature mRNA) can also cause amino acid substitutions.39 RNA editing in long noncoding RNAs can affect their secondary structure, abundance, stability, and interactions with miRNAs.40-46 Gong et al have created a database, LNCediting, to catalog the functional roles of RNA editing in long noncoding RNAs.47 

RNA editing in primary microRNAs (pri-miRNAs) typically inhibits miRNA maturation48-50 but can promote biogenesis in rare cases (eg, miR-197 and miR-203).51,52 Editing in the seed sequence alters target gene recognition53,54 and can also repress miRNA incorporation into RNA-induced silencing complex.55 RNA editing in Alu repeats of circular RNAs can unwind the dsRNA structure, promoting linear mRNA formation.56,57 In transfer RNAs (tRNAs), it enables wobble pairing for codon recognition.58 Pathogenic viral RNAs also exploit RNA editing to influence replication and pathogenesis,59 highlighting its role in diverse biological processes.

RNA editing can be quantified through various methods (Figure 2). Conventional approach analyze G/(G+A) or T/(T+C) peak height ratios in Sanger sequencing.60 The emergence of deep sequencing enables transcriptome-wide identification of RNA editing sites.61-64 Bulk-RNA sequencing, often using tools such as REDItools, maps the RNA editome across tissues.65 Microfluidics-based multiplex polymerase chain reaction (PCR) followed by deep sequencing is a targeted sequencing method developed to enable uniform and simultaneous amplification of nearly 1000 RNA editing loci for numerous samples.29,66 Single-cell RNA sequencing (scRNA-seq) data, although challenging due to lower quality and coverage, can identify RNA editing sites with high-depth, full-length data.67 Millions of RNA editing events have been curated and annotated in databases such as REDIportal,68 RADAR,69 and REDH,70 facilitating the exploration of the relevance between RNA editing loci, normal cellular processes, and associated disease pathogenesis.

Figure 2.

Detection methods for RNA editing events. (A) Sanger sequencing; detect the RNA editing levels by measuring ratios of G/(G+A) or T/(T+C) peaks. (B) Bulk-RNA sequencing; used to identify differences between RNA and DNA sequences, aided by REDItools suite. (C) mmPCR sequencing; uniformly and simultaneously amplify up to 960 loci in 48 samples, followed by sequencing. (D) Single-cell RNA sequencing; after preprocessing, alignment, UMI/barcode aggregation, and filtering steps, RNA editing sites can be annotated using tools such as ANNOVAR. (E) Digital PCR is a highly sensitive technique that allows for the detection of rare RNA editing events in hot spot regions by partitioning thousands of PCR reactions and using ≥2 fluorescent probes to determine the absolute proportions of sequence variants. (F) RESS-qPCR; detects wild-type (A) or edited (I) bases by incorporating mismatches in primers. Primer design strategy (i) uses the tetra-primer amplification refractory mutation system (ARMS) principles; strategy (ii) is used for positions that are incompatible with the Tetra-primer ARMS method due to significant differences in GC content upstream and downstream of the edited position. (G) Two-tailed qPCR for isomiRs. Specialized hemiprobes are used as primers for reverse transcription of both edited and unedited miRNAs. (H) LNA primer for isomiRs. This approach leverages tailored qPCR primers to detect miRNA isoforms with different melting temperatures, ensuring high sensitivity and accuracy in distinguishing miRNA variations. cDNA, complementary DNA; FW, forward; isomiRs, isoform of miRNAs; LNA, locked nucleic acid; mmPCR, microfluidics-based multiplex PCR; nt, nucleotides; pos, positive; RESS-qPCR, RNA editing site-specifc quantitative PCR; Rev, reverse; UMI, unique molecular identifier; wt, wide type.

Figure 2.

Detection methods for RNA editing events. (A) Sanger sequencing; detect the RNA editing levels by measuring ratios of G/(G+A) or T/(T+C) peaks. (B) Bulk-RNA sequencing; used to identify differences between RNA and DNA sequences, aided by REDItools suite. (C) mmPCR sequencing; uniformly and simultaneously amplify up to 960 loci in 48 samples, followed by sequencing. (D) Single-cell RNA sequencing; after preprocessing, alignment, UMI/barcode aggregation, and filtering steps, RNA editing sites can be annotated using tools such as ANNOVAR. (E) Digital PCR is a highly sensitive technique that allows for the detection of rare RNA editing events in hot spot regions by partitioning thousands of PCR reactions and using ≥2 fluorescent probes to determine the absolute proportions of sequence variants. (F) RESS-qPCR; detects wild-type (A) or edited (I) bases by incorporating mismatches in primers. Primer design strategy (i) uses the tetra-primer amplification refractory mutation system (ARMS) principles; strategy (ii) is used for positions that are incompatible with the Tetra-primer ARMS method due to significant differences in GC content upstream and downstream of the edited position. (G) Two-tailed qPCR for isomiRs. Specialized hemiprobes are used as primers for reverse transcription of both edited and unedited miRNAs. (H) LNA primer for isomiRs. This approach leverages tailored qPCR primers to detect miRNA isoforms with different melting temperatures, ensuring high sensitivity and accuracy in distinguishing miRNA variations. cDNA, complementary DNA; FW, forward; isomiRs, isoform of miRNAs; LNA, locked nucleic acid; mmPCR, microfluidics-based multiplex PCR; nt, nucleotides; pos, positive; RESS-qPCR, RNA editing site-specifc quantitative PCR; Rev, reverse; UMI, unique molecular identifier; wt, wide type.

Close modal

To validate these RNA editing sites, sensitive and cost-efficient strategies known as site-specific quantitative PCR (qPCR) and digital PCR have been developed and proven effective in clinical samples.32,71-75 Finally, specialized methods, such as 2-tailed reverse transcription qPCR (RT-qPCR) and RT-qPCR with locked nucleic acid primers, have been developed to analyze RNA editing in miRNAs.76-78 Together, this comprehensive tool kit for identifying and quantifying RNA editing provides researchers with unparalleled opportunities to gain in-depth insights into RNA biology and disease mechanisms. In the following sections, we focus on our explorations into the RNA editing landscape in the context of hematopoiesis.

There is now extensive evidence to suggest a role for RNA editing in hematopoiesis (Table 1). Jameson et al showed that lentiviral overexpression of ADAR1 p150 in cord blood cells skews differentiation toward the myeloid lineage79 and affects self-renewal and pluripotency genes.80 Wang et al81 published a seminal paper showing that ADAR1 haploinsufficiency leads to embryonic lethality before embryonic day 14 (E14), likely due in part to observed hematopoietic system abnormalities. Hartner et al82 also observed lethality around E12 using ADAR1-null mutant model, with liver disintegration and major defects in definitive hematopoiesis. It seems that widespread stress-induced apoptosis occurs in ADAR1 knockout mice.83 In the sections that follow, we discuss the role that ADAR1 plays during normal hematopoiesis.

Table 1.

ADAR1 knockout models and the impact on hematopoietic cell development

Cell typeGenetic modelRangeOutcomesReferences
Embryonic hematopoiesis Adar1+/− Genome wide Died before E14 with defects in the hematopoietic system 81  
Adar1−/− Genome wide Died around E12 with liver integration 82  
Embryonic
HSPCs 
Mx1-Cre; Adarf/− Hematopoietic system Result in low engraftment and elicit a global IFN response 84  
Adult HSPCs SCL-Cre-ER; Adarf/- HSCs Reduced LT-HSC (LKS+CD34lo) number and elevated IFN signaling 84  
Adult HSPCs MSCV-Cre; Adar1f/f
ER-Cre; Adar1f/f 
HSPCs Low engraftment but increased frequency of SLAM HSCs (LinCD48CD150+)
Increase apoptosis of differentiating HPCs (Linc-Kit+Sca-1
85  
Erythropoiesis Epor-Cre; Adar1Δ/−
Epor-Cre; Adar1Δ/E861A 
From Pre-Meg-E population downward Died in utero at E14.5
Macrocytic anemia and splenomegaly 
86  
Myeloid cells LysM-Cre; Adar1f/f Myelomonocytic cells Exhibit significantly reduced AM numbers, giant AM, and significant alveolar lipid accumulation 86,87  
Macrophages Lyz2(LysM)-Cre; Adarf/f Macrophages Establish an antitumor microenvironment 88  
DCs Cd11c-Cre; Adar1f/f DCs Fail to expand normal numbers of CD103+ DC
Increase inflammatory cDC2-like cells and promote early resistance against SARS-CoV-2 infection 
87,89  
T cells Lck-Cre; Adar1f/f Early T-cell stage (from DN stage onward) Reduce TCRβ expression but increase expression of type I ISGs in thymocytes 90,91  
T cells Cd4-Cre; Adar1f/f Late T-cell stage (from DP stage onward) Cause abnormal thymic T-cell maturation and autoimmunity 92  
T cells Foxp3-Cre; Adar1f/f Tregs Loss of peripheral Treg loss and autoimmune disorders 93  
B cells Cd19-Cre; Adar1f/f Late B-cell stage (from pre–B-cell stage onward) Reduce immature B and recirculating mature B cells; elevated apoptosis and ISGs 94,96  
B cells Mb1-Cre; Adar1f/f Early B-cell stage (from pre- to pro-B cells onward) Regulate pro–B- to large pre–B-cell transition and regulate pre-BCR expression to control large to small pre–B-cell transition 95  
B cells Mb1-Cre; Adar1f/f; MD4Tg/+ Drive B cell directly to BCR+ immature with a prearranged BCR transgene Display lower expression of IgM on immature B cells 95  
B cells Aicda-Cre; Adar1f/f Activated B cells Exhibit defective TD Ab response and diminished GC B cells 96  
Cell typeGenetic modelRangeOutcomesReferences
Embryonic hematopoiesis Adar1+/− Genome wide Died before E14 with defects in the hematopoietic system 81  
Adar1−/− Genome wide Died around E12 with liver integration 82  
Embryonic
HSPCs 
Mx1-Cre; Adarf/− Hematopoietic system Result in low engraftment and elicit a global IFN response 84  
Adult HSPCs SCL-Cre-ER; Adarf/- HSCs Reduced LT-HSC (LKS+CD34lo) number and elevated IFN signaling 84  
Adult HSPCs MSCV-Cre; Adar1f/f
ER-Cre; Adar1f/f 
HSPCs Low engraftment but increased frequency of SLAM HSCs (LinCD48CD150+)
Increase apoptosis of differentiating HPCs (Linc-Kit+Sca-1
85  
Erythropoiesis Epor-Cre; Adar1Δ/−
Epor-Cre; Adar1Δ/E861A 
From Pre-Meg-E population downward Died in utero at E14.5
Macrocytic anemia and splenomegaly 
86  
Myeloid cells LysM-Cre; Adar1f/f Myelomonocytic cells Exhibit significantly reduced AM numbers, giant AM, and significant alveolar lipid accumulation 86,87  
Macrophages Lyz2(LysM)-Cre; Adarf/f Macrophages Establish an antitumor microenvironment 88  
DCs Cd11c-Cre; Adar1f/f DCs Fail to expand normal numbers of CD103+ DC
Increase inflammatory cDC2-like cells and promote early resistance against SARS-CoV-2 infection 
87,89  
T cells Lck-Cre; Adar1f/f Early T-cell stage (from DN stage onward) Reduce TCRβ expression but increase expression of type I ISGs in thymocytes 90,91  
T cells Cd4-Cre; Adar1f/f Late T-cell stage (from DP stage onward) Cause abnormal thymic T-cell maturation and autoimmunity 92  
T cells Foxp3-Cre; Adar1f/f Tregs Loss of peripheral Treg loss and autoimmune disorders 93  
B cells Cd19-Cre; Adar1f/f Late B-cell stage (from pre–B-cell stage onward) Reduce immature B and recirculating mature B cells; elevated apoptosis and ISGs 94,96  
B cells Mb1-Cre; Adar1f/f Early B-cell stage (from pre- to pro-B cells onward) Regulate pro–B- to large pre–B-cell transition and regulate pre-BCR expression to control large to small pre–B-cell transition 95  
B cells Mb1-Cre; Adar1f/f; MD4Tg/+ Drive B cell directly to BCR+ immature with a prearranged BCR transgene Display lower expression of IgM on immature B cells 95  
B cells Aicda-Cre; Adar1f/f Activated B cells Exhibit defective TD Ab response and diminished GC B cells 96  

Ab, antibody; AM, alveolar macrophages; BCR, B-cell receptor; DP, double positive; GC, germinal center; HPC, hematopoietic progenitor cell; IgM, immunoglobulin M; LT-HSC, long term HSC; Pre-Meg-E, premegakaryocyte-erythroid; SARS-CoV-2, severe acute respiratory syndrome coronavirus 2; TCRβ, T-cell receptor beta chain; TD, T cell dependent; Treg, regulatory T cell.

HSPCs

Inspired by the gross animal-level effects of an ADAR1 knockout, researchers have conducted efforts to identify the cell-specific impact of ADAR1 ablation. For example, deletion of ADAR1 in fetal liver hematopoietic stem cells (HSCs) (Mx1-Cre-Adarf/−) resulted in notably low engraftment.84 ADAR1 deletion in adult HSCs (SCL-Cre-ER-Adarf/−) revealed that ADAR1 is essential for preventing the exhaustion of long-term HSCs (Linc-Kit+Sca-1+CD34lo) and IFN signaling.84 However, in our laboratory, using ER-Cre-Adar1f/f mice, we showed that ADAR1 plays a preferential role in promoting the proliferation of hematopoietic progenitor cells (Linc-Kit+Sca-1) compared with more primitive HSCs (Linc-Kit+Sca-1+).85 ADAR1-deficient HSCs successfully engrafted and were sustained for >6 months. This inconsistency may stem from variations in mouse models, gene excision efficiency, and the manipulation process.

Progeny of common myeloid progenitors

ADAR1 also plays a crucial role in the development of differentiated hematopoietic cells. Apart from its role in fetal erythropoiesis,81,86 ADAR1 knockout in Epor-Cre mice (Epor-Cre-Adar1Δ/−) induces macrocytic anemia or splenomegaly in adult, whereas an editing-deficient model (Epor-Cre-Adar1Δ/E861A) was underrepresented from around E15.5, indicative of a vital role of catalytic function of ADAR1 in murine erythropoiesis.86 Results from LysM-Cre-AdarΔ/Δ mice, however, suggested that ADAR1 was dispensable for myelopoiesis based on peripheral blood parameters.86 In contrast, others later demonstrated that specific deletion of ADAR1 in myeloid cells using LysM-Cre-AdarΔ/Δ mice led to significantly reduced numbers of alveolar macrophages.87 The same mouse model also proved that the loss of ADAR1 in macrophages promoted an antitumor microenvironment in combination with IFN-γ.88 In dendritic cells (DCs), CD11c-Cre-Adar1–/– mice exhibited impaired CD103+ DC expansion.87 Additionally, ADAR1-deficient antigen-presenting cells (CD11c-Cre+/−Adar1f/fR26YFP) showed an increased number of inflammatory type-2 conventional dendritic cells (cDC2)-like cells, elevated activated tissue-resident memory T cells, and an antiviral gene signature.89 

Lymphogenesis

Numerous studies have also investigated the role of ADAR1 in lymphocyte development (Table 1). Deletion of ADAR1 at the early T-cell stage (Lck-cre-Adar1f/f) resulted in severe thymic atrophy and impaired transition from DN3 (CD44-CD25+) to DN4 (CD44-CD25-) stage, along with reduced T-cell receptor β expression and increased expression of type I ISGs.90,91 Aberrant thymic T-cell maturation and excessive ISG expression were also observed in Cd4-Cre-Adar1f/f mice when Adar1 was deleted at the late T-cell stage.92 Luca et al93 demonstrated that deletion of Adar1(Foxp3-Cre-Adar1f/f) in T regulatory cells led to reduced numbers of peripheral T regulatory cells, which ultimately resulted in compromised regulatory functions and contributed to autoimmune disorder progression.

The ADAR1 p150 isoform has a critical role in B lymphopoiesis. Marcu-Malina et al94 showed that CD19-Cre–mediated Adar1 ablation (in late B-cell stage) led to a significant reduction in immature and mature B cells, primarily due to increased apoptosis. Focusing on early B lymphopoiesis, Chen et al95 used Mb1-Cre-Adar1f/f mice (from pre to pro B cells onward) to prove that ADAR1 regulates the transition from pro-B to large pre-B cells. In Mb1-Cre-Adar1f/fMD4Tg/+ mice, in which transgenic B-cell development bypasses the early B-cell stage, ADAR1 plays a critical role in regulating pre–B-cell receptor expression. This regulation is crucial for the transition from large pre-B cells to small pre-B cells. Li et al96 used Aicda-Cre-Adar1f/f mice, in which ADAR1 was specifically deleted in activated B cells, to investigate the role of ADAR1 in adaptive immunity. They discovered that ADAR1-deficient mice had impaired T-cell–dependent antibody responses and a significant reduction in germinal center B cells. In summary, studies demonstrate that ADAR1 plays a critical role in T and B lymphopoiesis across multiple stages.

Key RNA editing events

Certain key ADAR1-mediated RNA editing events play a pivotal role in regulating hematopoiesis (Table 2). A genome-wide analysis of ADAR1 RNA substrates in fetal liver during erythropoiesis revealed that 40% of RNA editing events occur in the long 3′ UTR of 3 genes: Klf1, Oip5, and Optn.13 These edited events generate inosine-uracil (I-U) mismatches and prevent the activation of inflammatory responses. While in editing-deficient knockin mutant mice (Adar1E861A/E861A), unedited transcripts can be bound by MDA5 and/or retinoic acid inducible gene I (RIG-I), leading to the activation of the cellular dsRNA response and immune cascades.

Table 2.

Site-specific RNA editing in normal hematopoiesis

Cell typeGeneEditing sitesMechanismsOutcomesReferences
NK cells TM7SF3, EIF3I, and RFX7 Not stated Not stated Probably involved in translation remodeling 106  
Monocytes/macrophages SDHB Exon Premature stop codon (R46X) Facilitate cellular adaptation to hypoxia 102,104  
HSPCs Klf1, Oip5, Optn 3′ UTR Generates multiple I-U mismatches that act to prevent MDA5 oligomerization Inhibit the cellular dsRNA response 13,86  
Azin1 Exon Recode 367th amino acid from serine to glycine Maintain HSCs’ functions 24  
Cog3, Taf1c, Rtkn, Igbp1, Rrp15, H19, Mri1 Exon, lnRNA, and 3′ UTR Not stated Not stated 24  
EIF2AK2 3′ UTR Disrupt the miRNA binding specificity of miR-23a and miR-23b Maintain cellular homeostasis during HSPCs’ differentiation 64  
Cell typeGeneEditing sitesMechanismsOutcomesReferences
NK cells TM7SF3, EIF3I, and RFX7 Not stated Not stated Probably involved in translation remodeling 106  
Monocytes/macrophages SDHB Exon Premature stop codon (R46X) Facilitate cellular adaptation to hypoxia 102,104  
HSPCs Klf1, Oip5, Optn 3′ UTR Generates multiple I-U mismatches that act to prevent MDA5 oligomerization Inhibit the cellular dsRNA response 13,86  
Azin1 Exon Recode 367th amino acid from serine to glycine Maintain HSCs’ functions 24  
Cog3, Taf1c, Rtkn, Igbp1, Rrp15, H19, Mri1 Exon, lnRNA, and 3′ UTR Not stated Not stated 24  
EIF2AK2 3′ UTR Disrupt the miRNA binding specificity of miR-23a and miR-23b Maintain cellular homeostasis during HSPCs’ differentiation 64  

NK cells, natural killer cells; lncRNA, long noncoding RNA.

Wang et al24 performed a comprehensive study of the RNA editome in murine hematopoietic cells. Their study identified 8 genes that undergo A-to-I RNA editing specifically in HSPCs, including Azin1, Cog3, Taf1c, Rtkn, Igbp1, Rrp15, H19, and Mri1. Among them, Azin1 can modulate HSC differentiation through interaction with Ddx1. Fully edited Azin1(S367G) enhances long-term hematopoietic reconstitution. Interestingly, Azin1 editing has been observed in patients with hepatocellular carcinoma and prostate cancer, in which it promotes a tumorigenic phenotype through mechanisms involving Azin1-Ddx1 nuclear translocation.30,97,98 This indicates that RNA editing at the same site can lead to divergent outcomes depending on the cell type.

Human HSPCs exhibit distinct RNA editing patterns that regulate lineage commitment and self-renewal. A study using single-cell RNA sequencing dataset identified several editing events linked to these processes.64 In EIF2AK2, editing at 4 miRNA binding sites in the 3′ UTR disrupts miR-23a/miR-23b binding, increasing EIF2AK2 expression and activating the stress response pathway. This mechanism initiates global translational attenuation as a protective mechanism to maintain cellular homeostasis during HSPC differentiation.64 RNA editing in genes such as CASP8 and ZBTB1 also plays a role in HSPC differentiation.

Given the critical role of RNA editing in normal hematopoiesis, the REDH database was developed to explore RNA editing events in both normal and abnormal hematopoiesis.70 

RNA editing facilitates cellular adaptation

RNA editing events not only maintain steady-state hematopoiesis but also facilitate cellular adaptation to environmental stimuli such as temperature changes, hypoxia, inflammation, and aging.99-101 For instance, APOBEC3A-mediated C-to-U editing occurs during M1 macrophage polarization and in monocytes exposed to hypoxia and IFN.102,103 In the SDHB gene, a rapid increase in the C136U editing event creates a premature stop codon (R46X), inactivating the SDHB protein and potentially enhancing monocyte and macrophage responses to low oxygen.102,104,105 

In natural killer cells and lymphocytes, hypoxic stress or reduced mitochondrial activity can trigger APOBEC3G-mediated RNA editing of multiple mRNA substrates, including TM7SF3, RPL10A, and RFX7, which may lead to translational reprogramming, altering protein expression profiles to respond to cellular stress.106 Beyond C-to-U editing, ADAR1-mediated A-to-I editing can also be triggered by external factors.99,107 Collectively, RNA editing helps cells better respond to environmental changes.

Aberrant RNA editing of specific transcripts has been implicated in various cancers, including hematologic malignancies.108-110 We believe that elevated ADAR1, particularly p150, in hematologic malignancies shares a common mechanism. Specifically, in disease progression, inflammatory cytokines (interleukin-3 [IL-3], IL-6, IFN, and tumor necrosis factor -α) activate the JAK/STAT pathway, promoting ADAR1 p150 and other ISG transcription. This mechanism is validated in blast crisis (BC) chronic myeloid leukemia (CML)50 and multiple myeloma (MM).111 In MM, ADAR1 is amplified on 1q21, explaining its increased levels. Additionally, the IL6R-STAT3-ADAR1112 and NOTCH/IL-6/ADAR1111 axes sustain ADAR1 expression. In this section, we highlight the oncogenic roles of ADAR and site-specific RNA editing events involved in hematologic cancers (Table 3).

Table 3.

Site-specific RNA editing events in hematologic malignancies

DiseaseGeneEditing sitesMechanismsOutcomesReferences
AML PTPN6 Intron Create aberrant splicing isoform Nonfunctional PTPN6 dysregulates its suppressor function on cKit signaling 33,114,115  
AML STAT3 Intron Affect alternative splicing and increase a shorter form STAT3β Promote transformation of pre-LSCs into LSCs that drive therapy-resistant sAML transformation 35,117  
AML COPA CDS I164V Inhibit the colony-forming of t(8:21) AML cells 31  
 COG3 CDS I635V 
AML SOCS2-AS1 LncRNA Not stated Correlated with low mRNA abundance and poor prognosis 120  
AML Pri-miR-142 Drosha cleavage site Edited pri-miRNA was degraded by Tudor-SN Inhibit mature miR-142 biogenesis 48  
CML Pri-let-7 DROSHA and DICER cleavage sites Upregulate LIN28B Promote LSC proliferation 50  
CML Pri-miR-26a DROSHA cleavage site Inhibit maturation of miR-26a Upregulate miR-26a target transcripts, which activates cell cycle–related gene CDKN1A 37  
CML MDM2 3′UTR Prevents miR-155 binding Increase MDM2 expression and repress the p53 tumor suppressor 37,38  
CML GSK3β Intron Increase a novel in-frame splice deletion Increase β-catenin expression and serial leukemia engraftment potential 79,121  
CLL miR-184, miR-589, miR-6503, miR3157 miRNAs Deregulate mRNA target network Affect the pathogenesis of CLL 129  
CLL COG3, CDK13, FLNB, BLCAP Nonsynonymous Protein recoding Probably contribute to treatment sensitivity in CLL 23  
T-ALL MAVS, IL17RA 3′UTR Not stated Low editing events are probably associated with loss of LIC stemness 130  
 MYB Intron Not stated 
DLBCL PRDM1 Exon-intron junction Premature translation termination PRDM1 inactivation and inhibition of terminal differentiation in DLBCL 34  
DLBCL MAVS 3′ UTR Increase MAVS protein expression levels Increase inflammation cascade and T-cell exhaustion 133  
PEL pri-miRNA-K12-4 Stem region and seed region Regulate miRNA biogenesis and target specificity Regulate viral life cycle of KSHV 134  
MM GLI1 Exon R701G, increase GLI1 transcriptional activity and decrease SUFU binding Activate Hedgehog pathway and lead to therapeutic resistance 32  
MM NEIL1 Exon K242R, change the lesion specificity of NEIL1 Display ineffective oxidative damage repair capacity and loss-of-TSG function properties 137  
MM MDM4, EIF2AK2 3′ UTR Interact with p53 and NF-κB Promote MM progression 142  
DiseaseGeneEditing sitesMechanismsOutcomesReferences
AML PTPN6 Intron Create aberrant splicing isoform Nonfunctional PTPN6 dysregulates its suppressor function on cKit signaling 33,114,115  
AML STAT3 Intron Affect alternative splicing and increase a shorter form STAT3β Promote transformation of pre-LSCs into LSCs that drive therapy-resistant sAML transformation 35,117  
AML COPA CDS I164V Inhibit the colony-forming of t(8:21) AML cells 31  
 COG3 CDS I635V 
AML SOCS2-AS1 LncRNA Not stated Correlated with low mRNA abundance and poor prognosis 120  
AML Pri-miR-142 Drosha cleavage site Edited pri-miRNA was degraded by Tudor-SN Inhibit mature miR-142 biogenesis 48  
CML Pri-let-7 DROSHA and DICER cleavage sites Upregulate LIN28B Promote LSC proliferation 50  
CML Pri-miR-26a DROSHA cleavage site Inhibit maturation of miR-26a Upregulate miR-26a target transcripts, which activates cell cycle–related gene CDKN1A 37  
CML MDM2 3′UTR Prevents miR-155 binding Increase MDM2 expression and repress the p53 tumor suppressor 37,38  
CML GSK3β Intron Increase a novel in-frame splice deletion Increase β-catenin expression and serial leukemia engraftment potential 79,121  
CLL miR-184, miR-589, miR-6503, miR3157 miRNAs Deregulate mRNA target network Affect the pathogenesis of CLL 129  
CLL COG3, CDK13, FLNB, BLCAP Nonsynonymous Protein recoding Probably contribute to treatment sensitivity in CLL 23  
T-ALL MAVS, IL17RA 3′UTR Not stated Low editing events are probably associated with loss of LIC stemness 130  
 MYB Intron Not stated 
DLBCL PRDM1 Exon-intron junction Premature translation termination PRDM1 inactivation and inhibition of terminal differentiation in DLBCL 34  
DLBCL MAVS 3′ UTR Increase MAVS protein expression levels Increase inflammation cascade and T-cell exhaustion 133  
PEL pri-miRNA-K12-4 Stem region and seed region Regulate miRNA biogenesis and target specificity Regulate viral life cycle of KSHV 134  
MM GLI1 Exon R701G, increase GLI1 transcriptional activity and decrease SUFU binding Activate Hedgehog pathway and lead to therapeutic resistance 32  
MM NEIL1 Exon K242R, change the lesion specificity of NEIL1 Display ineffective oxidative damage repair capacity and loss-of-TSG function properties 137  
MM MDM4, EIF2AK2 3′ UTR Interact with p53 and NF-κB Promote MM progression 142  

CDS, coding sequence; Drosha, mouse protein; DROSHA, human protein; LIC, leukemia-initiating cells; lncRNA, long noncoding RNA; PEL, primary effusion lymphoma; sAML, secondary acute myeloid leukemia; TSG, tumor suppressor gene.

AML

Several studies have explored the role of ADAR in acute myeloid leukemia (AML).31,113 In 2000, researchers found that protein tyrosine phosphatase nonreceptor type 6 (PTPN6), known for splicing mutations in hematopoietic disorders, undergoes RNA editing in AML. An A-to-I edit at position 7866 in intron 3 leads to an extended isoform. This hyperediting in AML may cause PTPN6 haploinsufficiency, weakening its tumor suppressor role in cKit signaling.33,114,115 Nishikura et al48 discovered that pri-miR-142-3p was the first microRNA edited by ADAR1 p110 and ADAR2, blocking its maturation. Another study116 reported miR-142 downregulation in patients with AML, indicating its potential as a prognostic marker. A study117 on RNA editing events during the transition from pre-LSCs to LSCs in AML showed that the bone marrow’s proinflammatory microenvironment increased ADAR1 p150 expression, leading to higher editing in STAT3 intron 21. This produced a shorter form of STAT3β isoform,35 which inhibits β-catenin degradation118 and helps LSCs maintain self-renewal.119 

Meduri et al120 analyzed The Cancer Genome Atlas (TCGA) AML data and found that SOCS2-AS1 editing is linked to poor survival negatively correlated with mRNA levels. They also observed reduced ADAR2 expression in AML, an unexpected phenomenon also confirmed in other studies.31 In this specific case, transcription of the ADAR2 gene is disrupted by the t(8;21) translocation. The reduction of ADAR2 levels results in the loss of RNA editing in mRNAs encoding components of the oligomeric Golgi complex 3 (Cog3) and coatomer subunit α (COPA). Overexpression of the edited forms inhibited the colony-forming ability of Kasumi-1 cells, suggesting ADAR2-mediated RNA editing plays a crucial role in preventing the development of therapy-resistant forms of core-binding factor AML.

CML

Jamieson et al79 first demonstrated the role of ADAR1 p150 in reprogramming malignant progenitor cells in CML. Subsequent studies have explored the involvement of ADAR1-mediated A-to-I editing in cell cycle transit, self-renewal, and the maintenance of LSCs. Overexpression of ADAR1 p150 induced myeloid skewing via PU.1 upregulation and created an aberrant spliced isoform (an in-frame splice deletion of exons 8 and 9) of glycogen synthase kinase 3β in CML progenitors, enhancing β-catenin expression and leukemia engraftment potential.121 Additionally, increased editing events in genes such as MDM2, APOBEC3D, GLI1, and AZIN172 have been identified and validated from chronic-phase progenitors to BC AML, which have been associated with stem cell fate122 and DNA mutagenesis.123 

Zipeto et al50 further proved the active role ADAR1 played in sustaining LSC self-renewal. Mechanistically, the activated JAK2/STAT and BCR-ABL1 (breakpoint cluster region-Abelson 1) signaling in CML converged on ADAR1 activation, mediating RNA editing of pri-let-7 miRNA family, thereby impairing their maturation. The reduced let-7 levels fail to target LIN28B, thus promoting self-renewal.124,125 Jiang et al37 found that editing at the DROSHA (human protein) cleavage site of pri-miR-26a inhibits miR-26a biogenesis, which normally suppresses CDKN1A via its target gene EZH2, thus promoting cell cycle progression and increasing self-renewal of CD34+ HSPCs. They also identified a ∼600-nucleotide region in the 3′ UTR of MDM2 that serves as a hot spot for RNA editing, where several miRNAs (such as miR-155) have their target sites. This editing increases MDM2 transcript abundance and accelerates the degradation of p53, ultimately promoting the transformation from chronic phase to BC CML.126 

CLL

In samples from patients with chronic lymphocytic leukemia (CLL), ADAR1 is presumed to interact with DROSHA, thereby inhibiting the biogenesis of pri-miR-16-1,127 a validated tumor suppressor in CLL.128 Notably, it is not ADAR1’s catalytic RNA-editing activity but rather its RNA-binding domain and nuclear localization domain that are responsible for affecting miRNA processing. Furthermore, a comprehensive analysis of miRNA editing in CLL and normal B cells identified 4 clinically relevant miRNAs that undergo robust RNA editing in CLL samples: miR-184, miR-589, miR-3157, and miR-6503. These edited miRNAs contributed to a dysregulated mRNA network.129 

In addition to the miRNA editome analysis, researchers conducted matched RNA sequencing and whole-exome sequencing of samples from patients with CLL. They identified 19 recurrent editing sites in genes such as COG3, CDK13, and COPA and revealed that RNA editing patterns are specific to CLL cohorts with different clinical outcomes. Moreover, ADAR knockout increased the sensitivity of CLL cell line MEC1 to treatments such as fludarabine and ibrutinib in vitro. These findings highlight ADAR as a promising target for future combination treatment strategies in CLL.23 

ALL

Jiang et al recently uncovered the critical role of ADAR1 in patients with relapsed T-cell acute lymphoblastic leukemia (T-ALL).130 ADAR1 maintained leukemia-initiating cell survival and self-renewal by mediating malignant A-to-I editing, which suppresses MDA5-directed dsRNA sensing. Loss of ADAR1 not only alleviated the leukemia burden in a xenograft model derived from patients with T-ALL but also reduced hyperediting events, such as those in the 3′ UTR of MAVS and IL17RA and in the intronic region of MYB.130 These findings suggest that targeting of ADAR1 could be a promising therapeutic strategy for eradicating T-ALL leukemia-initiating cells.

Lymphoma

RNA editing events have been linked to subtype-specific non-Hodgkin lymphoma regulation.131 Wayne et al34 first elucidated a potential role for RNA editing in lymphoma development via a mutational analysis of PRDM1. PRMD1 editing leads to its premature translation termination, which partially explains the pathogenesis of diffuse large B-cell lymphoma (DLBCL), given that PRDM1 is a well-known transcription suppressor participating in plasma cell differentiation.132 

Pecori et al133 studied ADAR1’s role in immune evasion in DLBCL by profiling genomic and transcriptomic data from 106 patient samples. They identified numerous edited or mutated genes involved in the apoptosis/p53 signaling pathway, NF-κB, and RIG-I–like receptor signaling pathways. An intriguing candidate is the RNA editing of MAVS 3′ UTR, which positively correlated with increased MAVS protein levels, inflammation cascade activation, and T-cell exhaustion, predicting poor prognosis in affected patients. Most excitingly, restoring MAVS editing via targeted base editing in ADAR-deficient RC-K8 cells (DLBCL-derived cell line) enhanced NF-κB and type I IFN signaling cascades.

RNA editing influences the life cycle of Kaposi sarcoma–associated herpesvirus (KSHV), the cause of primary effusion lymphoma. In this context, editing in the stem region of pri-miR-K12-4-3p affects miRNA maturation, whereas changes in the seed region alter target specificity, promoting more efficient KSHV infection. These findings deepen our understanding of KSHV-induced primary effusion lymphoma.134 

MM

A characteristic feature of MM is the amplification of chromosome 1q21, in which the ADAR1 gene is located.135,136 This amplification frequently leads to ADAR1 overexpression and is associated with increased RNA editing events.137 Notably, GLI1, a key Hedgehog (Hh) pathway activator, is edited in relapsed MM and plasma cell leukemia,32 enhancing Hh signaling and contributing to therapeutic resistance.138-140 Additionally, ADAR1 editing of NEIL1, a base-excision repair gene, boosts MM cell proliferation, reduces DNA repair, and increases drug resistance to standard MM therapies.131,141 

In a comparative analysis of whole-exome sequencing and RNA sequencing data, Tasakis et al142 further illustrated the dual role of ADAR1 in facilitating MM progression, which is driven by a combination of increased RNA events and the introduction of novel DNA mutations. Finally, among the edited cancer-related targets, Mdm4 and Eif2ak2 have significant roles in MM by interacting with the p53 and NF-κB pathways, respectively. Although these transcripts have been identified as edited in relevant databases, the specific mechanisms underlying the editing effects remain unclear.68,69 

Studies have also explored RNA editing patterns in pediatric hematologic malignancies and other blood cancers, although we will not delve into those details here.143 

Although the role of A-to-I editing in blood cancers is well documented, APOBEC-mediated C-to-U editing has also been implicated in malignancies. For instance, activation-induced cytidine deaminase (AID)-mediated mutations contribute to B-cell lymphoma, and overexpression of AID has been shown to induce T-lymphoma or B-leukemia/lymphoma.144,145 APOBEC3A overexpression has been associated with C-to-U editing of SF3B1 and KMT2A in hematologic neoplasms.146 Furthermore, APOBEC3B is upregulated in lymphoma cell lines,147 and APOBEC3G acts as a prosurvival factor that sensitizes lymphoma cells to radiation therapy.148 

As outlined in this review, growing evidence suggests that dysregulated ADAR activity and RNA editing events are involved in both hematopoiesis and abnormal hematopoiesis (Figure 3), particularly in cancer progression, therapy resistance, and poor prognosis.

Figure 3.

The role of RNA editing in hematopoietic system. An increasing body of research indicates various roles of RNA editing in the hematopoietic system. The most classical role of RNA editing involves regulating the immune response, specifically by adding inosine to dsRNAs to prevent the activation of the MDA5/MAVS signaling pathway, thereby avoiding the activation of IFN and ISGs. RNA editing is also crucial for maintaining hematopoietic homeostasis, particularly because a normal RNA editome ensures the proper function of HSPCs and guarantees normal lineage commitment. Additionally, in response to external stimuli such as hypoxia, temperature changes, or inflammation, RNA editing (primarily C-to-U) helps cells adapt. During cell aging, upregulated ADAR1 p150 can accelerate the aging of HSPCs, promote myeloid lineage skewing, and modulate inflammation-responsive transcription factors (TFs). Finally, in the case of abnormal hematopoiesis, under the influence of the inflammatory environment within the bone marrow niche, we find that tumor cells upregulate ADAR expression, leading to aberrant RNA editing events (such as Azin1, STAT3, MDM2, pri-let7, and pri-miR-26a). These altered RNA editing patterns cause dysregulation of the cell cycle and contribute to the proliferation of LSCs, promoting treatment resistance and poor prognosis. The human population and mouse icons represent the sources of the studied samples. TNF-α, tumor necrosis factor-α.

Figure 3.

The role of RNA editing in hematopoietic system. An increasing body of research indicates various roles of RNA editing in the hematopoietic system. The most classical role of RNA editing involves regulating the immune response, specifically by adding inosine to dsRNAs to prevent the activation of the MDA5/MAVS signaling pathway, thereby avoiding the activation of IFN and ISGs. RNA editing is also crucial for maintaining hematopoietic homeostasis, particularly because a normal RNA editome ensures the proper function of HSPCs and guarantees normal lineage commitment. Additionally, in response to external stimuli such as hypoxia, temperature changes, or inflammation, RNA editing (primarily C-to-U) helps cells adapt. During cell aging, upregulated ADAR1 p150 can accelerate the aging of HSPCs, promote myeloid lineage skewing, and modulate inflammation-responsive transcription factors (TFs). Finally, in the case of abnormal hematopoiesis, under the influence of the inflammatory environment within the bone marrow niche, we find that tumor cells upregulate ADAR expression, leading to aberrant RNA editing events (such as Azin1, STAT3, MDM2, pri-let7, and pri-miR-26a). These altered RNA editing patterns cause dysregulation of the cell cycle and contribute to the proliferation of LSCs, promoting treatment resistance and poor prognosis. The human population and mouse icons represent the sources of the studied samples. TNF-α, tumor necrosis factor-α.

Close modal

Indeed, numerous studies have shown that ADAR1 knockdown impairs the self-renewal capacity of malignant cells,50,130 and ADAR1 deletion can lead to regression of AML and CML in murine models149,150 or treatment sensitivity in CLL cell lines.23 These findings open avenues into potential treatment approaches that might involve (1) the selective inhibition of ADAR activity or (2) the prevention of dysregulated A-to-I editing at specific sites. We discuss these 2 potential approaches in the following sections.

ADAR inhibitors

Despite considerable efforts, there are currently no US Food and Drug Administration–approved drugs targeting ADAR. For example, adenosine analogs such as 8-azaadenosine (8-aza-A) and 8-chloroadenosine (8-Cl-A) can suppress ADAR activity in various cancer contexts.50,151 However, results showed that these inhibitors are neither selective for ADAR nor effective in inhibiting A-to-I RNA editing of ADAR substrates.152 ZYS-1 is a newly synthesized small molecule that targets the deaminase activity of ADAR1, showing promising antitumor efficacy and favorable safety for patients with prostate cancer.153 It may be worthwhile to explore the application of ZYS-1 in hematologic malignancies. Another promising inhibitor, Rebecsinib, has shown potential in treating high-risk myelofibrosis and secondary AML. It prevents the expression of the ADAR1 p150 splice isoform and blocks malignant A-to-I editing–mediated LSC self-renewal, while sparing normal HSPCs.73 Additionally, research is ongoing into strategies for inhibiting ADAR deaminase activity,154 marking this as a promising therapeutic area. RNA editing inhibition may treat blood malignancies, but side effects on hematopoiesis and immunity must be carefully considered, with an optimized therapeutic window balancing efficacy and safety.

RNA editing transcript inhibitors

Antisense oligonucleotide (ASO) therapy, designed to block RNA editing by targeting complementary sequences, shows promise for correcting disease-causing mutations. Chemically modified oligonucleotides enhance stability, binding affinity, specificity, and reduce nuclease cleavage and immune response. For instance, ASOs with 2’-O-methyl/locked nucleic acid modifications have shown efficacy in inhibiting NEIL1 editing.155 Meanwhile, ASO3.2 has been shown to abolish AZIN1 editing without affecting splicing and translation and inhibits cancer cell viability in vitro or in patient-derived xenograft models.156 Progress in this area is restrained, however, given the promiscuous RNA editing events that occur in cancer context; halting disease progression by targeting only a few specific RNA editing sites remains unlikely. Consequently, further studies are needed on the application of site-specific RNA editing inhibitors.

Diagnostic potential of RNA editing

RNA editing holds significant potential for clinical diagnostics, with studies exploring its application in diseases such as neurological disorders and cancers.157 Efforts to diagnose hematologic malignancies have focused on detecting miRNA editing in plasma and extracellular vesicles from patients with myelodysplastic syndrome.158 A study found increased RNA editing in early-stage myelodysplastic syndrome, but only a few miRNAs showed edited forms with low rates of A-to-I changes, highlighting the need for improved detection sensitivity.

ADARs, with their ability to bind dsRNA and perform A-to-I deamination, have potential therapeutic applications.159 Recent studies have explored by the use of antisense guide RNAs to harness endogenous ADARs160-163 or delivering exogenous ADARs for targeted RNA editing.162,164 Although DNA editing can provide durable and potentially permanent cures for certain diseases,165 RNA-targeted modifications without genome integration offer flexible and temporary changes, making them safer for conditions requiring transient effects, such as acute pain, viral infections, obesity, and inflammation.166 

To further explore the therapeutic potential of RNA editing, we queried the ClinVar website and identified nearly 14 000 G>A missense and nonsense mutations among pathogenic single-nucleotide variants to date, which represent prime candidates for targeting and correction to the wild-type sequence using ADAR-mediated RNA editing therapy.166,167 Of course, the candidate mutations are not limited to G>A mutations, given the redundancy of codons in the genetic code. This flexibility allows for various types of nucleotide substitutions to be targeted for therapeutic correction, as demonstrated by successful practices in the treatment of inherited retinal diseases.168 

In addition to their RNA editing–dependent effects, ADARs can also exert RNA editing–independent functions by binding to DNA or interacting with proteins involving DORSHA, DICER1, and HuR.56 Furthermore, the Zα domain of ADAR1 can bind to the G-quadruplex in the c-Myc promoter.169 Given the critical role of c-Myc in regulating HSPC function and hematopoietic malignancies,170 this interaction warrants further investigation.

Future research targeting RNA editing enzymes or specific editing events holds promise for advancing cancer treatment. We envisage RNA editing therapy as a promising tool for correcting disease-causing mutations by leveraging endogenous or engineered ADAR enzymes. First, we need to overcome limitations in delivery systems, target specificity, and off-target effects and take time to explore the molecular mechanisms governing RNA editing efficiency and accuracy to better understand how to safely and effectively harness this technology for therapeutic applications.

This work was supported by the Ministry of Science and Technology of China (grant number 2021YFA1100900), the National Natural Science Foundation of China (grant numbers 92468206 and 92368202), the Chinese Academy of Medical Sciences (CAMS) Innovation Fund for Medical Sciences (grant numbers 2021-I2M-1-040 and 2021-I2M-1-019), the CAMS Fundamental Research Funds for Central Research Institutes (grant number 3332021093), and the Haihe Laboratory of Cell Ecosystem Innovation Fund (grant number 24HHXBSS00006).

Contribution: S.P. wrote the initial draft of the manuscript and conducted the literature review; and H.C. and T.C. provided revisions and helped refine the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Hui Cheng, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Rd, Heping District, Tianjin 300020, China; email: chenghui@ihcams.ac.cn; and Tao Cheng, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 288 Nanjing Rd, Heping District, Tianjin 300020, China; email: chengtao@ihcams.ac.cn.

1.
Cappannini
A
,
Ray
A
,
Purta
E
, et al
.
MODOMICS: a database of RNA modifications and related information. 2023 update
.
Nucleic Acids Res
.
2024
;
52
(
D1
):
D239
-
D244
.
2.
Nishikura
K
.
Functions and regulation of RNA editing by ADAR deaminases
.
Annu Rev Biochem
.
2010
;
79
(
1
):
321
-
349
.
3.
Conticello
SG
.
The AID/APOBEC family of nucleic acid mutators
.
Genome Biol
.
2008
;
9
(
6
):
229
.
4.
Keegan
LP
,
Leroy
A
,
Sproul
D
,
O’Connell
MA
.
Adenosine deaminases acting on RNA (ADARs): RNA-editing enzymes
.
Genome Biol
.
2004
;
5
(
2
):
209
.
5.
Kim
U
,
Wang
Y
,
Sanford
T
,
Zeng
Y
,
Nishikura
K
.
Molecular cloning of cDNA for double-stranded RNA adenosine deaminase, a candidate enzyme for nuclear RNA editing
.
Proc Natl Acad Sci U S A
.
1994
;
91
(
24
):
11457
-
11461
.
6.
Melcher
T
,
Maas
S
,
Herb
A
,
Sprengel
R
,
Higuchi
M
,
Seeburg
PH
.
RED2, a brain-specific member of the RNA-specific adenosine deaminase family
.
J Biol Chem
.
1996
;
271
(
50
):
31795
-
31798
.
7.
Gerber
A
,
O’Connell
MA
,
Keller
W
.
Two forms of human double-stranded RNA-specific editase 1 (hRED1) generated by the insertion of an Alu cassette
.
RNA
.
1997
;
3
(
5
):
453
-
463
.
8.
Chen
CX
,
Cho
DS
,
Wang
Q
,
Lai
F
,
Carter
KC
,
Nishikura
K
.
A third member of the RNA-specific adenosine deaminase gene family, ADAR3, contains both single- and double-stranded RNA binding domains
.
RNA
.
2000
;
6
(
5
):
755
-
767
.
9.
George
CX
,
Samuel
CE
.
Characterization of the 5’-flanking region of the human RNA-specific adenosine deaminase ADAR1 gene and identification of an interferon-inducible ADAR1 promoter
.
Gene
.
1999
;
229
(
1-2
):
203
-
213
.
10.
Fritz
J
,
Strehblow
A
,
Taschner
A
,
Schopoff
S
,
Pasierbek
P
,
Jantsch
MF
.
RNA-regulated interaction of transportin-1 and exportin-5 with the double-stranded RNA-binding domain regulates nucleocytoplasmic shuttling of ADAR1
.
Mol Cell Biol
.
2009
;
29
(
6
):
1487
-
1497
.
11.
Fisher
AJ
,
Beal
PA
.
Structural perspectives on adenosine to inosine RNA editing by ADARs
.
Mol Ther Nucleic Acids
.
2024
;
35
(
3
):
102284
.
12.
Hu
S-B
,
Heraud-Farlow
J
,
Sun
T
, et al
.
ADAR1p150 prevents MDA5 and PKR activation via distinct mechanisms to avert fatal autoinflammation
.
Mol Cell
.
2023
;
83
(
21
):
3869
-
3884.e7
.
13.
Liddicoat
BJ
,
Piskol
R
,
Chalk
A
, et al
.
RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself
.
Science
.
2015
;
349
(
6252
):
1115
-
1120
.
14.
Xu
L-D
,
Öhman
M
.
ADAR1 editing and its role in cancer
.
Genes
.
2018
;
10
(
1
):
12
.
15.
Frezza
V
,
Chellini
L
,
Del Verme
A
,
Paronetto
MP
.
RNA editing in cancer progression
.
Cancers
.
2023
;
15
(
21
):
5277
.
16.
Herbert
A
.
ADAR and immune silencing in cancer
.
Trends Cancer
.
2019
;
5
(
5
):
272
-
282
.
17.
Maurano
M
,
Snyder
J
,
Connelly
C
,
Henao-Mejia
J
,
Sidrauski
C
,
Stetson
DB
.
Protein kinase R and the integrated stress response drive immunopathology caused by mutations in the RNA deaminase ADAR1
.
Immunity
.
2021
;
54
(
9
):
1948
-
1960.e5
.
18.
Jiang
Q
,
Crews
LA
,
Holm
F
,
Jamieson
CHM
.
RNA editing-dependent epitranscriptome diversity in cancer stem cells
.
Nat Rev Cancer
.
2017
;
17
(
6
):
381
-
392
.
19.
Lu
D
,
Lu
J
,
Liu
Q
,
Zhang
Q
.
Emerging role of the RNA-editing enzyme ADAR1 in stem cell fate and function
.
Biomark Res
.
2023
;
11
(
1
):
61
.
20.
Minervini
CF
,
Parciante
E
,
Impera
L
, et al
.
Epitranscriptomics in normal and malignant hematopoiesis
.
Int J Mol Sci
.
2020
;
21
(
18
):
6578
.
21.
Rosselló-Tortella
M
,
Ferrer
G
,
Esteller
M
.
Epitranscriptomics in hematopoiesis and hematologic malignancies
.
Blood Cancer Discov
.
2020
;
1
(
1
):
26
-
31
.
22.
Ahmad
H.M.
. Role of post-transcriptional gene regulation in hematological malignancies. In:
Jain
B.P.
,
Goswami
S.K.
,
Sharma
T.
, eds.
Post-transcriptional Gene Regulation in Human Disease
. 1st ed..
Academic Press
;
2022
:
105
-
117
.
23.
Gassner
FJ
,
Zaborsky
N
,
Buchumenski
I
, et al
.
RNA editing contributes to epitranscriptome diversity in chronic lymphocytic leukemia
.
Leukemia
.
2021
;
35
(
4
):
1053
-
1063
.
24.
Wang
F
,
He
J
,
Liu
S
, et al
.
A comprehensive RNA editome reveals that edited Azin1 partners with DDX1 to enable hematopoietic stem cell differentiation
.
Blood
.
2021
;
138
(
20
):
1939
-
1952
.
25.
Teoh
PJ
,
Koh
MY
,
Chng
WJ
.
ADARs, RNA editing and more in hematological malignancies
.
Leukemia
.
2021
;
35
(
2
):
346
-
359
.
26.
Nishikura
K
.
A-to-I editing of coding and non-coding RNAs by ADARs
.
Nat Rev Mol Cell Biol
.
2016
;
17
(
2
):
83
-
96
.
27.
Song
Y
,
Yang
W
,
Fu
Q
, et al
.
irCLASH reveals RNA substrates recognized by human ADARs
.
Nat Struct Mol Biol
.
2020
;
27
(
4
):
351
-
362
.
28.
Basilio
C
,
Wahba
AJ
,
Lengyel
P
,
Speyer
JF
,
Ochoa
S
.
Synthetic polynucleotides and the amino acid code, V.
.
Proc Natl Acad Sci U S A
.
1962
;
48
(
4
):
613
-
616
.
29.
Tan
MH
,
Li
Q
,
Shanmugam
R
, et al
.
Dynamic landscape and regulation of RNA editing in mammals
.
Nature
.
2017
;
550
(
7675
):
249
-
254
.
30.
Chen
L
,
Li
Y
,
Lin
CH
, et al
.
Recoding RNA editing of AZIN1 predisposes to hepatocellular carcinoma
.
Nat Med
.
2013
;
19
(
2
):
209
-
216
.
31.
Guo
M
,
Chan
THM
,
Zhou
Q
, et al
.
Core-binding factor fusion downregulation of ADAR2 RNA editing contributes to AML leukemogenesis
.
Blood
.
2023
;
141
(
25
):
3078
-
3090
.
32.
Lazzari
E
,
Mondala
PK
,
Santos
ND
, et al
.
Alu-dependent RNA editing of GLI1 promotes malignant regeneration in multiple myeloma
.
Nat Commun
.
2017
;
8
(
1
):
1922
.
33.
Beghini
A
,
Ripamonti
CB
,
Peterlongo
P
, et al
.
RNA hyperediting and alternative splicing of hematopoietic cell phosphatase (PTPN6) gene in acute myeloid leukemia
.
Hum Mol Genet
.
2000
;
9
(
15
):
2297
-
2304
.
34.
Tam
W
,
Gomez
M
,
Chadburn
A
,
Lee
JW
,
Chan
WC
,
Knowles
DM
.
Mutational analysis of PRDM1 indicates a tumor-suppressor role in diffuse large B-cell lymphomas
.
Blood
.
2006
;
107
(
10
):
4090
-
4100
.
35.
Goldberg
L
,
Abutbul-Amitai
M
,
Paret
G
,
Nevo-Caspi
Y
.
Alternative splicing of STAT3 is affected by RNA editing
.
DNA Cell Biol
.
2017
;
36
(
5
):
367
-
376
.
36.
Hsiao
Y-HE
,
Bahn
JH
,
Yang
Y
, et al
.
RNA editing in nascent RNA affects pre-mRNA splicing
.
Genome Res
.
2018
;
28
(
6
):
812
-
823
.
37.
Jiang
Q
,
Isquith
J
,
Zipeto
MA
, et al
.
Hyper-editing of cell-cycle regulatory and tumor suppressor RNA promotes malignant progenitor propagation
.
Cancer Cell
.
2019
;
35
(
1
):
81
-
94.e7
.
38.
Zhang
L
,
Yang
C-S
,
Varelas
X
,
Monti
S
.
Altered RNA editing in 3′ UTR perturbs microRNA-mediated regulation of oncogenes and tumor-suppressors
.
Sci Rep
.
2016
;
6
(
1
):
23226
.
39.
Xing
Y
,
Nakahama
T
,
Wu
Y
, et al
.
RNA editing of AZIN1 coding sites is catalyzed by ADAR1 p150 after splicing
.
J Biol Chem
.
2023
;
299
(
7
):
104840
.
40.
Picardi
E
,
D’Erchia
AM
,
Gallo
A
,
Montalvo
A
,
Pesole
G
.
Uncovering RNA editing sites in long non-coding RNAs
.
Front Bioeng Biotechnol
.
2014
;
2
:
64
.
41.
Goldstein
B
,
Agranat-Tamir
L
,
Light
D
,
Ben-Naim Zgayer
O
,
Fishman
A
,
Lamm
AT
.
A-to-I RNA editing promotes developmental stage–specific gene and lncRNA expression
.
Genome Res
.
2017
;
27
(
3
):
462
-
470
.
42.
Vlachogiannis
NI
,
Sachse
M
,
Georgiopoulos
G
, et al
.
Adenosine-to-inosine Alu RNA editing controls the stability of the pro-inflammatory long noncoding RNA NEAT1 in atherosclerotic cardiovascular disease
.
J Mol Cell Cardiol
.
2021
;
160
:
111
-
120
.
43.
Stellos
K
,
Gatsiou
A
,
Stamatelopoulos
K
, et al
.
Adenosine-to-inosine RNA editing controls cathepsin S expression in atherosclerosis by enabling HuR-mediated post-transcriptional regulation
.
Nat Med
.
2016
;
22
(
10
):
1140
-
1150
.
44.
Silvestris
DA
,
Scopa
C
,
Hanchi
S
,
Locatelli
F
,
Gallo
A
.
De novo A-to-I RNA editing discovery in lncRNA
.
Cancers
.
2020
;
12
(
10
):
2959
.
45.
Chattopadhyay
P
,
Mehta
P
,
Kanika
, et al
.
RNA editing in host lncRNAs as potential modulator in SARS-CoV-2 variants-host immune response dynamics
.
iScience
.
2024
;
27
(
6
):
109846
.
46.
Qiu
J
,
Ma
X
,
Zeng
F
,
Yan
J
.
RNA editing regulates lncRNA splicing in human early embryo development
.
PLoS Comput Biol
.
2021
;
17
(
12
):
e1009630
.
47.
Gong
J
,
Liu
C
,
Liu
W
, et al
.
LNCediting: a database for functional effects of RNA editing in lncRNAs
.
Nucleic Acids Res
.
2017
;
45
(
D1
):
D79
-
D84
.
48.
Yang
W
,
Chendrimada
TP
,
Wang
Q
, et al
.
Modulation of microRNA processing and expression through RNA editing by ADAR deaminases
.
Nat Struct Mol Biol
.
2006
;
13
(
1
):
13
-
21
.
49.
Kawahara
Y
,
Megraw
M
,
Kreider
E
, et al
.
Frequency and fate of microRNA editing in human brain
.
Nucleic Acids Res
.
2008
;
36
(
16
):
5270
-
5280
.
50.
Zipeto
MA
,
Court
AC
,
Sadarangani
A
, et al
.
ADAR1 activation drives leukemia stem cell self-renewal by impairing let-7 biogenesis
.
Cell Stem Cell
.
2016
;
19
(
2
):
177
-
191
.
51.
Ota
H
,
Sakurai
M
,
Gupta
R
, et al
.
ADAR1 forms a complex with dicer to promote microRNA processing and RNA-induced gene silencing
.
Cell
.
2013
;
153
(
3
):
575
-
589
.
52.
Correia De Sousa
M
,
Gjorgjieva
M
,
Dolicka
D
,
Sobolewski
C
,
Foti
M
.
Deciphering miRNAs’ action through miRNA editing
.
IJMS
.
2019
;
20
(
24
):
6249
.
53.
Kawahara
Y
,
Zinshteyn
B
,
Sethupathy
P
,
Iizasa
H
,
Hatzigeorgiou
AG
,
Nishikura
K
.
Redirection of silencing targets by adenosine-to-inosine editing of miRNAs
.
Science, New Series
.
2007
;
315
(
5815
):
1137
-
1140
.
54.
Paul
D
,
Sinha
AN
,
Ray
A
, et al
.
A-to-I editing in human miRNAs is enriched in seed sequence, influenced by sequence contexts and significantly hypoedited in glioblastoma multiforme
.
Sci Rep
.
2017
;
7
(
1
):
2466
.
55.
Iizasa
H
,
Wulff
B-E
,
Alla
NR
, et al
.
Editing of Epstein-Barr virus-encoded BART6 microRNAs controls their dicer targeting and consequently affects viral latency
.
J Biol Chem
.
2010
;
285
(
43
):
33358
-
33370
.
56.
Daniel
C
,
Lagergren
J
,
Öhman
M
.
RNA editing of non-coding RNA and its role in gene regulation
.
Biochimie
.
2015
;
117
:
22
-
27
.
57.
Ivanov
A
,
Memczak
S
,
Wyler
E
, et al
.
Analysis of intron sequences reveals hallmarks of circular RNA biogenesis in animals
.
Cell Rep
.
2015
;
10
(
2
):
170
-
177
.
58.
Alazami
AM
,
Hijazi
H
,
Al-Dosari
MS
, et al
.
Mutation in ADAT3, encoding adenosine deaminase acting on transfer RNA, causes intellectual disability and strabismus
.
J Med Genet
.
2013
;
50
(
7
):
425
-
430
.
59.
Kim
K
,
Calabrese
P
,
Wang
S
, et al
.
The roles of APOBEC-mediated RNA editing in SARS-CoV-2 mutations, replication and fitness
.
Sci Rep
.
2022
;
12
(
1
):
14972
.
60.
Jepson
JEC
,
Reenan
RA
.
Genetic approaches to studying adenosine-to-inosine RNA editing
.
Methods Enzymol
.
2007
;
424
:
265
-
287
.
61.
Li
M
,
Wang
IX
,
Li
Y
, et al
.
Widespread RNA and DNA sequence differences in the human transcriptome
.
Science
.
2011
;
333
(
6038
):
53
-
58
.
62.
Blanc
V
,
Park
E
,
Schaefer
S
, et al
.
Genome-wide identification and functional analysis of Apobec-1-mediated C-to-U RNA editing in mouse small intestine and liver
.
Genome Biol
.
2014
;
15
(
6
):
R79
.
63.
Picardi
E
,
Manzari
C
,
Mastropasqua
F
,
Aiello
I
,
D'Erchia
AM
,
Pesole
G
.
Profiling RNA editing in human tissues: towards the inosinome Atlas
.
Sci Rep
.
2015
;
5
(
1
):
14941
.
64.
Wu
Y
,
Hao
S
,
Xu
X
, et al
.
A novel computational method enables RNA editome profiling during human hematopoiesis from scRNA-seq data
.
Sci Rep
.
2023
;
13
(
1
):
10335
.
65.
Picardi
E
,
Pesole
G
.
REDItools: high-throughput RNA editing detection made easy
.
Bioinformatics
.
2013
;
29
(
14
):
1813
-
1814
.
66.
Zhang
R
,
Li
X
,
Ramaswami
G
, et al
.
Quantifying RNA allelic ratios by microfluidic multiplex PCR and sequencing
.
Nat Methods
.
2014
;
11
(
1
):
51
-
54
.
67.
Lv
T
,
Jiang
S
,
Wang
X
,
Hou
Y
.
Profiling A-to-I RNA editing during mouse somatic reprogramming at the single-cell level
.
Heliyon
.
2023
;
9
(
7
):
e18133
.
68.
Mansi
L
,
Tangaro
MA
,
Lo Giudice
C
, et al
.
REDIportal: millions of novel A-to-I RNA editing events from thousands of RNAseq experiments
.
Nucleic Acids Res
.
2021
;
49
(
D1
):
D1012
-
D1019
.
69.
Ramaswami
G
,
Li
JB
.
RADAR: a rigorously annotated database of A-to-I RNA editing
.
Nucleic Acids Res
.
2014
;
42
(
Database issue
):
D109
-
D113
.
70.
Xu
J
,
He
J
,
Yang
J
, et al
.
REDH: A database of RNA editome in hematopoietic differentiation and malignancy
.
Chin Med J
.
2024
;
137
(
3
):
283
-
293
.
71.
Ye
S
,
Dhillon
S
,
Ke
X
,
Collins
AR
,
Day
IN
.
An efficient procedure for genotyping single nucleotide polymorphisms
.
Nucleic Acids Res
.
2001
;
29
(
17
):
E88
.
72.
Crews
LA
,
Jiang
Q
,
Zipeto
MA
, et al
.
An RNA editing fingerprint of cancer stem cell reprogramming
.
J Transl Med
.
2015
;
13
(
1
):
52
.
73.
Crews
LA
,
Ma
W
,
Ladel
L
, et al
.
Reversal of malignant ADAR1 splice isoform switching with Rebecsinib
.
Cell Stem Cell
.
2023
;
30
(
3
):
250
-
263.e6
.
74.
Jalili
P
,
Bowen
D
,
Langenbucher
A
, et al
.
Quantification of ongoing APOBEC3A activity in tumor cells by monitoring RNA editing at hotspots
.
Nat Commun
.
2020
;
11
(
1
):
2971
.
75.
Oh
S
,
Buisson
R
.
A digital PCR-based protocol to detect and quantify RNA editing events at hotspots
.
STAR Protoc
.
2022
;
3
(
1
):
101148
.
76.
Voss
G
,
Ceder
Y
.
Two-tailed RT-qPCR for the quantification of A-to-I-edited microRNA isoforms
.
Curr Protoc
.
2023
;
3
(
1
):
e645
.
77.
Voss
G
,
Edsjö
A
,
Bjartell
A
,
Ceder
Y
.
Quantification of microRNA editing using two-tailed RT-qPCR for improved biomarker discovery
.
RNA
.
2021
;
27
(
11
):
1412
-
1424
.
78.
Widmark
A
,
Sagredo
EA
,
Karlström
V
, et al
.
ADAR1- and ADAR2-mediated regulation of maturation and targeting of miR-376b to modulate GABA neurotransmitter catabolism
.
J Biol Chem
.
2022
;
298
(
3
):
101682
.
79.
Jiang
Q
,
Crews
L
,
Barrett
CL
, et al
.
ADAR1 promotes malignant progenitor reprogramming in chronic myeloid leukemia
.
Proc Natl Acad Sci U S A
.
2013
;
110
(
3
):
1041
-
1046
.
80.
Jiang
Q
,
Zipeto
MA
,
Delos Santos
N
, et al
.
RNA editing enzyme ADAR1 accelerates normal hematopoiesis cell cycle by regulating microRNA biogenesis
.
Blood
.
2016
;
128
(
22
):
887
.
81.
Wang
Q
,
Khillan
J
,
Gadue
P
,
Nishikura
K
.
Requirement of the RNA editing deaminase ADAR1 gene for embryonic erythropoiesis
.
Science
.
2000
;
290
(
5497
):
1765
-
1768
.
82.
Hartner
JC
,
Schmittwolf
C
,
Kispert
A
,
Müller
AM
,
Higuchi
M
,
Seeburg
PH
.
Liver disintegration in the mouse embryo caused by deficiency in the RNA-editing enzyme ADAR1
.
J Biol Chem
.
2004
;
279
(
6
):
4894
-
4902
.
83.
Wang
Q
,
Miyakoda
M
,
Yang
W
, et al
.
Stress-induced apoptosis associated with null mutation of ADAR1 RNA editing deaminase gene
.
J Biol Chem
.
2004
;
279
(
6
):
4952
-
4961
.
84.
Hartner
JC
,
Walkley
CR
,
Lu
J
,
Orkin
SH
.
ADAR1 is essential for the maintenance of hematopoiesis and suppression of interferon signaling
.
Nat Immunol
.
2009
;
10
(
1
):
109
-
115
.
85.
XuFeng
R
,
Boyer
MJ
,
Shen
H
, et al
.
ADAR1 is required for hematopoietic progenitor cell survival via RNA editing
.
Proc Natl Acad Sci U S A
.
2009
;
106
(
42
):
17763
-
17768
.
86.
Liddicoat
BJ
,
Hartner
JC
,
Piskol
R
, et al
.
Adenosine-to-inosine RNA editing by ADAR1 is essential for normal murine erythropoiesis
.
Exp Hematol
.
2016
;
44
(
10
):
947
-
963
.
87.
Baal
N
,
Cunningham
S
,
Obermann
H-L
, et al
.
ADAR1 is required for dendritic cell subset homeostasis and alveolar macrophage function
.
J Immunol
.
2019
;
202
(
4
):
1099
-
1111
.
88.
Lin
W
,
Luo
Y
,
Wu
J
, et al
.
Loss of ADAR1 in macrophages in combination with interferon gamma suppresses tumor growth by remodeling the tumor microenvironment
.
J Immunother Cancer
.
2023
;
11
(
11
):
e007402
.
89.
Adamska
JZ
,
Verma
R
,
Gupta
S
, et al
.
Ablation of Adar1 in myeloid cells imprints a global antiviral state in the lung and heightens early immunity against SARS-CoV-2
.
Cell Rep
.
2023
;
42
(
1
):
112038
.
90.
Xufeng
R
,
Nie
D
,
Yang
Q
,
Wang
W
,
Cheng
T
,
Wang
Q
.
RNA editing enzyme ADAR1 is required for early T cell development
.
Blood Sci
.
2020
;
2
(
1
):
27
-
32
.
91.
Vongpipatana
T
,
Nakahama
T
,
Shibuya
T
,
Kato
Y
,
Kawahara
Y
.
ADAR1 regulates early T cell development via MDA5-dependent and -independent pathways
.
J Immunol
.
2020
;
204
(
8
):
2156
-
2168
.
92.
Nakahama
T
,
Kato
Y
,
Kim
JI
, et al
.
ADAR1-mediated RNA editing is required for thymic self-tolerance and inhibition of autoimmunity
.
EMBO Rep
.
2018
;
19
(
12
):
e46303
.
93.
Luca
D
,
Lee
S
,
Hirota
K
, et al
.
Aberrant RNA sensing in regulatory T cells causes systemic autoimmunity
.
Sci Adv
.
2024
;
10
(
9
):
eadk0820
.
94.
Marcu-Malina
V
,
Goldberg
S
,
Vax
E
,
Amariglio
N
,
Goldstein
I
,
Rechavi
G
.
ADAR1 is vital for B cell lineage development in the mouse bone marrow
.
Oncotarget
.
2016
;
7
(
34
):
54370
-
54379
.
95.
Chen
W
,
Li
Y
,
Ruan
G-X
, et al
.
Adenosine deaminase acting on RNA-1 is essential for early B lymphopoiesis
.
Cell Rep
.
2022
;
41
(
8
):
111687
.
96.
Li
Y
,
Ruan
G-X
,
Chen
W
, et al
.
RNA-editing enzyme ADAR1 p150 isoform is critical for germinal center B cell response
.
J Immunol
.
2022
;
209
(
6
):
1071
-
1082
.
97.
Rossetti
C
,
Picardi
E
,
Ye
M
, et al
.
RNA editing signature during myeloid leukemia cell differentiation
.
Leukemia
.
2017
;
31
(
12
):
2824
-
2832
.
98.
Ghalali
A
,
Wang
L
,
Stopsack
KH
, et al
.
AZIN1 RNA editing alters protein interactions, leading to nuclear translocation and worse outcomes in prostate cancer
.
Exp Mol Med
.
2022
;
54
(
10
):
1713
-
1726
.
99.
Cayir
A
.
RNA A-to-I editing, environmental exposure, and human diseases
.
Crit Rev Toxicol
.
2021
;
51
(
5
):
456
-
466
.
100.
Van Der Werf I, Sneifer J, Jamieson C. RNA modifications shape hematopoietic stem cell aging: beyond the code
.
FEBS Lett
.
2024
;
598
(
22
):
2774
-
2775
.
101.
Ma
W
,
Pham
J
,
Klacking
EE
, et al
.
Detection and prevention of ADAR1p150-induced hematopoietic stem and progenitor cell aging
.
Blood
.
2023
;
142
(
suppl 1
):
4054
.
102.
Sharma
S
,
Patnaik
SK
,
Taggart
RT
, et al
.
APOBEC3A cytidine deaminase induces RNA editing in monocytes and macrophages
.
Nat Commun
.
2015
;
6
(
1
):
6881
.
103.
Alqassim
EY
,
Sharma
S
,
Khan
ANMNH
, et al
.
RNA editing enzyme APOBEC3A promotes pro-inflammatory M1 macrophage polarization
.
Commun Biol
.
2021
;
4
(
1
):
102
.
104.
Baysal
BE
,
De Jong
K
,
Liu
B
, et al
.
Hypoxia-inducible C-to-U coding RNA editing downregulates SDHB in monocytes
.
PeerJ
.
2013
;
1
:
e152
.
105.
Strehl
C
,
Fangradt
M
,
Fearon
U
,
Gaber
T
,
Buttgereit
F
,
Veale
DJ
.
Hypoxia: how does the monocyte-macrophage system respond to changes in oxygen availability?
.
J Leukoc Biol
.
2014
;
95
(
2
):
233
-
241
.
106.
Sharma
S
,
Wang
J
,
Alqassim
E
, et al
.
Mitochondrial hypoxic stress induces widespread RNA editing by APOBEC3G in natural killer cells
.
Genome Biol
.
2019
;
20
(
1
):
37
.
107.
Nevo-Caspi
Y
,
Amariglio
N
,
Rechavi
G
,
Paret
G
.
A-to-I RNA editing is induced upon hypoxia
.
Shock
.
2011
;
35
(
6
):
585
-
589
.
108.
Dominissini
D
,
Moshitch-Moshkovitz
S
,
Amariglio
N
,
Rechavi
G
.
Adenosine-to-inosine RNA editing meets cancer
.
Carcinogenesis
.
2011
;
32
(
11
):
1569
-
1577
.
109.
Paz
N
,
Levanon
EY
,
Amariglio
N
, et al
.
Altered adenosine-to-inosine RNA editing in human cancer
.
Genome Res
.
2007
;
17
(
11
):
1586
-
1595
.
110.
Han
L
,
Diao
L
,
Yu
S
, et al
.
The genomic landscape and clinical relevance of A-to-I RNA editing in human cancers
.
Cancer Cell
.
2015
;
28
(
4
):
515
-
528
.
111.
Lazzari
E.
.
Mechanisms of Drug-Escape and Eelapse in Multiple Myeloma: Interplay Between ADAR1 and NOTCH in Aberrant RNA Editing. Dissertation
.
Università degli Studi di Milano
;
2015
.
112.
Teoh
PJ
,
Chung
T-H
,
Chng
PYZ
,
Toh
SHM
,
Chng
WJ
.
IL6R-STAT3-ADAR1 (P150) interplay promotes oncogenicity in multiple myeloma with 1q21 amplification
.
Haematologica
.
2020
;
105
(
5
):
1391
-
1404
.
113.
Xiao
H
,
Cheng
Q
,
Wu
X
,
Tang
Y
,
Liu
J
,
Li
X
.
ADAR1 may be involved in the proliferation of acute myeloid leukemia cells via regulation of the Wnt pathway
.
Cancer Manag Res
.
2019
;
11
:
8547
-
8555
.
114.
Quelen
C
,
Eloit
Y
,
Noirot
C
,
Bousquet
M
,
Brousset
P
.
RNA editing in acute myeloid leukaemia with normal karyotype
.
Br J Haematol
.
2016
;
173
(
5
):
788
-
790
.
115.
Zipeto
MA
,
Jiang
Q
,
Melese
E
,
Jamieson
CHM
.
RNA rewriting, recoding, and rewiring in human disease
.
Trends Mol Med
.
2015
;
21
(
9
):
549
-
559
.
116.
Wang
F
,
Wang
X-S
,
Yang
G-H
, et al
.
miR-29a and miR-142-3p downregulation and diagnostic implication in human acute myeloid leukemia
.
Mol Biol Rep
.
2012
;
39
(
3
):
2713
-
2722
.
117.
Jiang
Q
,
Isquith
J
,
Ladel
L
, et al
.
Inflammation-driven deaminase deregulation fuels human pre-leukemia stem cell evolution
.
Cell Rep
.
2021
;
34
(
4
):
108670
.
118.
Wu
J
,
Keng
VW
,
Patmore
DM
, et al
.
Insertional mutagenesis identifies a STAT3/Arid1b/β-catenin pathway driving neurofibroma initiation
.
Cell Rep
.
2016
;
14
(
8
):
1979
-
1990
.
119.
Jamieson
CHM
,
Ailles
LE
,
Dylla
SJ
, et al
.
Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML
.
N Engl J Med
.
2004
;
351
(
7
):
657
-
667
.
120.
Meduri
E
,
Breeze
C
,
Marando
L
,
Richardson
SE
,
Huntly
BJP
.
The RNA editing landscape in acute myeloid leukemia reveals associations with disease mutations and clinical outcome
.
iScience
.
2022
;
25
(
12
):
105622
.
121.
Abrahamsson
AE
,
Geron
I
,
Gotlib
J
, et al
.
Glycogen synthase kinase 3beta missplicing contributes to leukemia stem cell generation
.
Proc Natl Acad Sci U S A
.
2009
;
106
(
10
):
3925
-
3929
.
122.
Ladel
L
,
Jamieson
C
.
Upping the antizyme: AZIN1 directs stem cell fate
.
Blood
.
2021
;
138
(
20
):
1910
-
1911
.
123.
Roberts
SA
,
Lawrence
MS
,
Klimczak
LJ
, et al
.
An APOBEC cytidine deaminase mutagenesis pattern is widespread in human cancers
.
Nat Genet
.
2013
;
45
(
9
):
970
-
976
.
124.
Copley
MR
,
Babovic
S
,
Benz
C
, et al
.
The Lin28b-let-7-Hmga2 axis determines the higher self-renewal potential of fetal haematopoietic stem cells
.
Nat Cell Biol
.
2013
;
15
(
8
):
916
-
925
.
125.
Piskounova
E
,
Polytarchou
C
,
Thornton
JE
, et al
.
Lin28A and Lin28B inhibit let-7 microRNA biogenesis by distinct mechanisms
.
Cell
.
2011
;
147
(
5
):
1066
-
1079
.
126.
Trotta
R
,
Vignudelli
T
,
Candini
O
, et al
.
BCR/ABL activates mdm2 mRNA translation via the La antigen
.
Cancer Cell
.
2003
;
3
(
2
):
145
-
160
.
127.
Allegra
D
,
Bilan
V
,
Garding
A
, et al
.
Defective DROSHA processing contributes to downregulation of MiR-15/-16 in chronic lymphocytic leukemia
.
Leukemia
.
2014
;
28
(
1
):
98
-
107
.
128.
Klein
U
,
Lia
M
,
Crespo
M
, et al
.
The DLEU2/miR-15a/16-1 cluster controls B cell proliferation and its deletion leads to chronic lymphocytic leukemia
.
Cancer Cell
.
2010
;
17
(
1
):
28
-
40
.
129.
Gassner
FJ
,
Zaborsky
N
,
Feldbacher
D
,
Greil
R
,
Geisberger
R
.
RNA editing alters miRNA function in chronic lymphocytic leukemia
.
Cancers
.
2020
;
12
(
5
):
1159
.
130.
Rivera
M
,
Zhang
H
,
Pham
J
, et al
.
Malignant A-to-I RNA editing by ADAR1 drives T cell acute lymphoblastic leukemia relapse via attenuating dsRNA sensing
.
Cell Rep
.
2024
;
43
(
2
):
113704
.
131.
Chen
C
,
Bundschuh
R
.
A-to-I editing Is subtype-specific in non-Hodgkin lymphomas
.
Genes
.
2024
;
15
(
7
):
864
.
132.
Xia
Y
,
Xu-Monette
ZY
,
Tzankov
A
, et al
.
Loss of PRDM1/BLIMP-1 function contributes to poor prognosis of activated B-cell-like diffuse large B-cell lymphoma
.
Leukemia
.
2017
;
31
(
3
):
625
-
636
.
133.
Pecori
R
,
Ren
W
,
Pirmoradian
M
, et al
.
ADAR1-mediated RNA editing promotes B cell lymphomagenesis
.
iScience
.
2023
;
26
(
6
):
106864
.
134.
Rajendren
S
,
Ye
X
,
Dunker
W
,
Richardson
A
,
Karijolich
J
.
The cellular and KSHV A-to-I RNA editome in primary effusion lymphoma and its role in the viral lifecycle
.
Nat Commun
.
2023
;
14
(
1
):
1367
.
135.
Terpos
E
,
Eleutherakis-Papaiakovou
V
,
Dimopoulos
M-A
.
Clinical implications of chromosomal abnormalities in multiple myeloma
.
Leuk Lymphoma
.
2006
;
47
(
5
):
803
-
814
.
136.
Chng
WJ
,
Glebov
O
,
Bergsagel
PL
,
Kuehl
WM
.
Genetic events in the pathogenesis of multiple myeloma
.
Best Pract Res Clin Haematol
.
2007
;
20
(
4
):
571
-
596
.
137.
Teoh
PJ
,
An
O
,
Chung
T-H
, et al
.
Aberrant hyperediting of the myeloma transcriptome by ADAR1 confers oncogenicity and is a marker of poor prognosis
.
Blood
.
2018
;
132
(
12
):
1304
-
1317
.
138.
Shimokawa
T
,
Rahman
MF-U
,
Tostar
U
, et al
.
RNA editing of the GLI1 transcription factor modulates the output of Hedgehog signaling
.
RNA Biol
.
2013
;
10
(
2
):
321
-
333
.
139.
Peacock
CD
,
Wang
Q
,
Gesell
GS
, et al
.
Hedgehog signaling maintains a tumor stem cell compartment in multiple myeloma
.
Proc Natl Acad Sci U S A
.
2007
;
104
(
10
):
4048
-
4053
.
140.
Blotta
S
,
Jakubikova
J
,
Calimeri
T
, et al
.
Canonical and noncanonical Hedgehog pathway in the pathogenesis of multiple myeloma
.
Blood
.
2012
;
120
(
25
):
5002
-
5013
.
141.
Yeo
J
,
Goodman
RA
,
Schirle
NT
,
David
SS
,
Beal
PA
.
RNA editing changes the lesion specificity for the DNA repair enzyme NEIL1
.
Proc Natl Acad Sci U S A
.
2010
;
107
(
48
):
20715
-
20719
.
142.
Tasakis
RN
,
Laganà
A
,
Stamkopoulou
D
, et al
.
ADAR1 can drive multiple myeloma progression by acting both as an RNA editor of specific transcripts and as a DNA mutator of their cognate genes
.
bioRxiv
.
Preprint posted online 21 June 2020
.
143.
Wen
J
,
Rusch
M
,
Brady
SW
, et al
.
The landscape of coding RNA editing events in pediatric cancer
.
BMC Cancer
.
2021
;
21
(
1
):
1233
.
144.
Komeno
Y
,
Kitaura
J
,
Watanabe-Okochi
N
, et al
.
AID-induced T-lymphoma or B-leukemia/lymphoma in a mouse BMT model
.
Leukemia
.
2010
;
24
(
5
):
1018
-
1024
.
145.
Pasqualucci
L
,
Bhagat
G
,
Jankovic
M
, et al
.
AID is required for germinal center-derived lymphomagenesis
.
Nat Genet
.
2008
;
40
(
1
):
108
-
112
.
146.
Sharma
S
,
Patnaik
SK
,
Kemer
Z
,
Baysal
BE
.
Transient overexpression of exogenous APOBEC3A causes C-to-U RNA editing of thousands of genes
.
RNA Biol
.
2017
;
14
(
5
):
603
-
610
.
147.
Shinohara
M
,
Io
K
,
Shindo
K
, et al
.
APOBEC3B can impair genomic stability by inducing base substitutions in genomic DNA in human cells
.
Sci Rep
.
2012
;
2
:
806
.
148.
Nowarski
R
,
Wilner
OI
,
Cheshin
O
, et al
.
APOBEC3G enhances lymphoma cell radioresistance by promoting cytidine deaminase-dependent DNA repair
.
Blood
.
2012
;
120
(
2
):
366
-
375
.
149.
Steinman
RA
,
Yang
Q
,
Gasparetto
M
, et al
.
Deletion of the RNA-editing enzyme ADAR1 causes regression of established chronic myelogenous leukemia in mice
.
Int J Cancer
.
2013
;
132
(
8
):
1741
-
1750
.
150.
Peng
L
,
Yang
X
,
Zhang
Y
, et al
.
Effect of ADAR1 on the development of MLL-AF9 induced murine AML
.
Zhonghua Xue Ye Xue Za Zhi
.
2015
;
36
(
5
):
383
-
388
.
151.
Ramírez-Moya
J
,
Baker
AR
,
Slack
FJ
,
Santisteban
P
.
ADAR1-mediated RNA editing is a novel oncogenic process in thyroid cancer and regulates miR-200 activity
.
Oncogene
.
2020
;
39
(
18
):
3738
-
3753
.
152.
Cottrell
KA
,
Torres
LS
,
Dizon
MG
,
Weber
JD
.
8-Azaadenosine and 8-chloroadenosine are not selective inhibitors of ADAR
.
Cancer Res Commun
.
2021
;
1
(
2
):
56
-
64
.
153.
Wang
X
,
Li
J
,
Zhu
Y
, et al
.
Targeting ADAR1 with a novel small-molecule for the treatment of prostate cancer
.
Research Square
.
Preprint posted online 21 September 2021
.
154.
Rehwinkel
J
,
Mehdipour
P
.
ADAR1: from basic mechanisms to inhibitors
.
Trends in Cell Biology
.
2025
;
35
(
1
):
59
-
73
.
155.
Mizrahi
RA
,
Schirle
NT
,
Beal
PA
.
Potent and selective inhibition of A-to-I RNA editing with 2’-O-methyl/locked nucleic acid-containing antisense oligoribonucleotides
.
ACS Chem Biol
.
2013
;
8
(
4
):
832
-
839
.
156.
Tay
DJT
,
Song
Y
,
Peng
B
, et al
.
Targeting RNA editing of antizyme inhibitor 1: a potential oligonucleotide-based antisense therapy for cancer
.
Mol Ther
.
2021
;
29
(
11
):
3258
-
3273
.
157.
Salvetat
N
,
Checa-Robles
FJ
,
Patel
V
, et al
.
A game changer for bipolar disorder diagnosis using RNA editing-based biomarkers
.
Transl Psychiatry
.
2022
;
12
(
1
):
182
.
158.
Hrustincova
A
,
Krejcik
Z
,
Kundrat
D
, et al
.
Circulating small noncoding RNAs have specific expression patterns in plasma and extracellular vesicles in myelodysplastic syndromes and are predictive of patient outcome
.
Cells
.
2020
;
9
(
4
):
794
.
159.
Woolf
TM
,
Chase
JM
,
Stinchcomb
DT
.
Toward the therapeutic editing of mutated RNA sequences
.
Proc Natl Acad Sci U S A
.
1995
;
92
(
18
):
8298
-
8302
.
160.
Merkle
T
,
Merz
S
,
Reautschnig
P
, et al
.
Precise RNA editing by recruiting endogenous ADARs with antisense oligonucleotides
.
Nat Biotechnol
.
2019
;
37
(
2
):
133
-
138
.
161.
Katrekar
D
,
Yen
J
,
Xiang
Y
, et al
.
Efficient in vitro and in vivo RNA editing via recruitment of endogenous ADARs using circular guide RNAs
.
Nat Biotechnol
.
2022
;
40
(
6
):
938
-
945
.
162.
Rauch
S
,
He
E
,
Srienc
M
,
Zhou
H
,
Zhang
Z
,
Dickinson
BC
.
Programmable RNA-guided RNA effector proteins built from human parts
.
Cell
.
2019
;
178
(
1
):
122
-
134.e12
.
163.
Yi
Z
,
Qu
L
,
Tang
H
, et al
.
Engineered circular ADAR-recruiting RNAs increase the efficiency and fidelity of RNA editing in vitro and in vivo
.
Nat Biotechnol
.
2022
;
40
(
6
):
946
-
955
.
164.
Cox
DBT
,
Gootenberg
JS
,
Abudayyeh
OO
, et al
.
RNA editing with CRISPR-Cas13
.
Science
.
2017
;
358
(
6366
):
1019
-
1027
.
165.
Li
H
,
Yang
Y
,
Hong
W
,
Huang
M
,
Wu
M
,
Zhao
X
.
Applications of genome editing technology in the targeted therapy of human diseases: mechanisms, advances and prospects
.
Sig Transduct Target Ther
.
2020
;
5
(
1
):
1
-
23
.
166.
Booth
BJ
,
Nourreddine
S
,
Katrekar
D
, et al
.
RNA editing: Expanding the potential of RNA therapeutics
.
Mol Ther
.
2023
;
31
(
6
):
1533
-
1549
.
167.
Diaz Quiroz
JF
,
Siskel
LD
,
Rosenthal
JJC
.
Site-directed A → I RNA editing as a therapeutic tool: moving beyond genetic mutations
.
RNA
.
2023
;
29
(
4
):
498
-
505
.
168.
Schneider
N
,
Steinberg
R
,
Ben-David
A
, et al
.
A pipeline for identifying guide RNA sequences that promote RNA editing of nonsense mutations that cause inherited retinal diseases
.
Mol Ther Nucleic Acids
.
2024
;
35
(
1
):
102130
.
169.
Kang
H-J
,
Le
TVT
,
Kim
K
,
Hur
J
,
Kim
KK
,
Park
HJ
.
Novel interaction of the Z-DNA binding domain of human ADAR1 with the oncogenic c-Myc promoter G-quadruplex
.
J Mol Biol
.
2014
;
426
(
14
):
2594
-
2604
.
170.
Ahmadi
SE
,
Rahimi
S
,
Zarandi
B
,
Chegeni
R
,
Safa
M
.
MYC: a multipurpose oncogene with prognostic and therapeutic implications in blood malignancies
.
J Hematol Oncol
.
2021
;
14
(
1
):
121
.
Sign in via your Institution