rna editing rna editing by adar1 prevents mda5 sensing of ... · rna editing rna editing by adar1...

7
RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself Brian J. Liddicoat, 1,2 Robert Piskol, 3 Alistair M. Chalk, 1,2 Gokul Ramaswami, 3 Miyoko Higuchi, 4 Jochen C. Hartner, 5 Jin Billy Li, 3 * Peter H. Seeburg, 4 * Carl R. Walkley 1,2 *Adenosine-to-inosine (A-to-I) editing is a highly prevalent posttranscriptional modification of RNA, mediated by ADAR (adenosine deaminase acting on RNA) enzymes. In addition to RNA editing, additional functions have been proposed for ADAR1.To determine the specific role of RNA editing by ADAR1, we generated mice with an editing-deficient knock-in mutation (Adar1 E861A , where E861A denotes Glu 861 Ala 861 ). Adar1 E861A/E861A embryos died at ~E13.5 (embryonic day 13.5), with activated interferon and double-stranded RNA (dsRNA)sensing pathways. Genome-wide analysis of the in vivo substrates of ADAR1 identified clustered hyperediting within long dsRNA stem loops within 3untranslated regions of endogenous transcripts. Finally, embryonic death and phenotypes of Adar1 E861A/E861A were rescued by concurrent deletion of the cytosolic sensor of dsRNA, MDA5. A-to-I editing of endogenous dsRNA is the essential function of ADAR1, preventing the activation of the cytosolic dsRNA response by endogenous transcripts. A denosine-to-inosine (A-to-I) editing is the most prevalent form of RNA base modifi- cation in mammals. Hundreds of thousands of A-to-I editing events have been reported in the human transcriptome (13). A-to-I editing is catalyzed by ADAR (adenosine deam- inase acting on RNA) enzymes, which deaminate genomically encoded A-to-I in double-stranded RNA (dsRNA). A-to-I editing predominantly oc- curs in noncoding, repetitive elements such as inverted Alus and short interspersed nuclear ele- ments (SINEs) (3, 4). There are three mamma- lian ADAR proteins: ADAR1, -2, and -3. ADAR1 is widely expressed during embryonic and post- natal development and is present as a predom- inantly nuclear, constitutive ADAR1p110 isoform expressed in all tissues and an additional in- terferon (IFN)inducible ADAR1p150 isoform that is found in both the nucleus and the cyto- plasm (5). Adar1 -/- (null for both isoforms) and Adar1p150 -/- mice die in utero at E11.5 (embry- onic day 11.5) to E12.5, due to failed erythropoi- esis and fetal liver (FL) disintegration (68). Only ADAR1 and ADAR2 demonstrate editing activity in vitro. The AMPA receptor GluA2 pre- mRNA is edited by ADAR2 in the brain, con- verting a glutamine residue to an arginine in a functionally critical position (9). Adar2 -/- mice die from seizures and were rescued by the genomic substitution of the single edited aden- osine for guanosine, mimicking editing of the transcript (10). In contrast to this elegant single- substrate paradigm that describes the Adar2 -/- phenotype, no such editing site(s) described to date have resolved the physiological requirement for ADAR1. To directly determine the contribution of A-to-I editing to ADAR1s biological function, we generated a constitutive knock-in of an editing- deficient ADAR1 allele in mice (Adar1 E861A , where E861A denotes Glu 861 Ala 861 ) (Fig. 1A and fig. S1A), homologous to the human ADAR1 E912A allele, which is catalytically inactive in vitro (11) (also see supplementary materials and methods). In E12.5 whole embryos, ADAR1p110 protein was detected in Adar1 E861A/E861A samples at compa- rable levels to Adar1 +/+ and Adar1 E861A/+ controls (Fig. 1B). Additionally, Adar1 E861A/E861A embryos had elevated expression of ADAR1p150 (Fig. 1B). Adar1 E861A/+ heterozygous mice were normal, were present at the expected Mendelian ratio, and had no discernible phenotype. No viable Adar1 E861A/E861A pups were born from Adar1 E861A/+ intercrosses and were found to be dying at ~E13.5 (Fig. 1C). Adar1 E861A/E861A yolk sacs were pale, and embryos appeared developmentally delayed compared with controls (Fig. 1D), despite being normal at E12.5 (fig. S1, D and E). At E13.5, Adar1 E861A/E861A FL was smaller, with eightfold fewer viable cells (Fig. 1D and fig. S1E). We observed a failure in erythropoiesis with a severe loss of erythroblast populations (Fig. 1, E and F) and increased cell death (Fig. 1G), also apparent in other hematopoietic populations (fig. S2). Consistent with our previous analysis of the ADAR1 conditional allele (12), adult hemato- poietic stem cells (HSCs) expressing only the catalytically inactive ADAR1 could not be main- tained in vivo and were lost over time (fig. S3). The A-to-I editing activity of ADAR1 is essential for embryonic development and the maintenance of hematopoiesis in vivo. We performed transcriptional profiling on three independent littermate Adar1 +/+ and Adar1 E861A/E861A FLs. An absence of ADAR1 edit- ing resulted in the up-regulation of 383 transcripts (log 2 FC > 2), 258 of which are IFN-stimulated genes (ISGs) (Fig. 2A and table S1). Pathway anal- ysis revealed a profound enrichment of signatures activated after IFN treatment or viral infection (Fig. 2B and table S1). Of the 50 most differentially expressed genes, 44 were up-regulated by either type I IFN, type II IFN, or both (Fig. 2C and table S1). The derepression of ISGs was confirmed by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) (Fig. 2D). Further- more, we demonstrated that the transcriptional response in Adar1 E861A/E861A FLs closely parallels that of complete ADAR1 deficiency (12) (fig. S4A). Synthetic dsRNA sequences based on endoge- nous RNA containing adenosine but not inosine stimulate ISGs and apoptosis in vitro by binding to MDA5 and RIG-I (13). We defined a gene set associated with the response to IU-dsRNA (inosine- uracilpaired dsRNA) in human cells (13). Gene signatures of both Adar1 E861A/E861A FL and Adar1 -/ - HSCs were highly enriched for the IU-dsRNA response (Fig. 2E), demonstrating a species- conserved response to dsRNA that is constrained by the presence of inosine residues. Therefore, the deamination of adenosine in dsRNA by ADAR1 is necessary for suppression of the IFN response under homeostatic conditions. Fur- thermore, it suggested that the absence of A-to-I substitutions in endogenous dsRNA may initiate this response. To define ADAR1-specific editing events in vivo, we analyzed A-to-I (G) mismatches within the RNA sequencing data. Across all samples, 6167 A-to-I editing sites were identified: 5540 known (14) and 627 previously undiscovered (Fig. 3A). Strain-specific single-nucleotide polymorphisms were excluded, and only A-to-I mismatch fre- quencies that differed significantly between geno- types were considered (Fig. 3B). Using these criteria, 673 A-to-I editing sites were defined (Fig. 3A). Of these, 666 sites had reduced editing in mutants, including hyperedited loci, which had no detectable editing (Fig. 3B and tables S2 and S3). This confirmed that the Adar1 E861A allele is catalytically inactive and the majority of FL A- to-I editing is ADAR1 dependent. We validated 281 of the identified A-to-I sites on independent Adar1 E861A/E861A and Adar1 +/+ FL samples (15). All tested sites were confirmed as differentially edited in both FL and mouse embryonic fibro- blasts (MEFs) (table S4). Sanger sequencing validated ADAR1-specific Blcap Y2C , Mad2l1, Rbbp4, and Klf1 editing sites from independent FL sam- ples (Fig. 3C). ADAR1 is required for erythropoiesis (Fig. 1, E to G). Interestingly, 40% (264 of 666) of the ADAR1-specific editing sites were within hyper- edited clusters of only three genesKlf1, Oip5, and Optnwhich have peak or restricted expression in SCIENCE sciencemag.org 4 SEPTEMBER 2015 VOL 349 ISSUE 6252 1115 1 St. Vincents Institute of Medical Research, Fitzroy, Victoria 3065, Australia. 2 Department of Medicine, St. Vincents Hospital, University of Melbourne, Fitzroy, Victoria 3065, Australia. 3 Department of Genetics, Stanford University, Stanford, CA 94305, USA. 4 Department of Molecular Neurobiology, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany. 5 Taconic Biosciences, 51063 Cologne, Germany. *These authors contributed equally to this work. Corresponding author. E-mail: [email protected] RESEARCH | REPORTS on November 12, 2020 http://science.sciencemag.org/ Downloaded from

Upload: others

Post on 12-Aug-2020

12 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of ... · RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself Brian J. Liddicoat,1,2 Robert

RNA EDITING

RNA editing by ADAR1 preventsMDA5 sensing of endogenous dsRNAas nonselfBrian J. Liddicoat,1,2 Robert Piskol,3 Alistair M. Chalk,1,2 Gokul Ramaswami,3

Miyoko Higuchi,4 Jochen C. Hartner,5 Jin Billy Li,3*Peter H. Seeburg,4* Carl R. Walkley1,2*†

Adenosine-to-inosine (A-to-I) editing is a highly prevalent posttranscriptional modification ofRNA, mediated by ADAR (adenosine deaminase acting on RNA) enzymes. In addition to RNAediting, additional functions have been proposed for ADAR1.To determine the specific role ofRNA editing by ADAR1, we generated mice with an editing-deficient knock-in mutation(Adar1E861A, where E861A denotes Glu861→Ala861). Adar1E861A/E861A embryos died at ~E13.5(embryonic day 13.5), with activated interferon and double-stranded RNA (dsRNA)–sensingpathways. Genome-wide analysis of the in vivo substrates of ADAR1 identified clusteredhyperediting within long dsRNA stem loops within 3′ untranslated regions of endogenoustranscripts. Finally, embryonic death and phenotypes of Adar1E861A/E861Awere rescued byconcurrent deletion of the cytosolic sensor of dsRNA, MDA5. A-to-I editing of endogenousdsRNA is the essential function of ADAR1, preventing the activation of the cytosolic dsRNAresponse by endogenous transcripts.

Adenosine-to-inosine (A-to-I) editing is themost prevalent form of RNA base modifi-cation in mammals. Hundreds of thousandsof A-to-I editing events have been reportedin the human transcriptome (1–3). A-to-I

editing is catalyzed by ADAR (adenosine deam-inase acting on RNA) enzymes, which deaminategenomically encoded A-to-I in double-strandedRNA (dsRNA). A-to-I editing predominantly oc-curs in noncoding, repetitive elements such asinverted Alus and short interspersed nuclear ele-ments (SINEs) (3, 4). There are three mamma-lian ADAR proteins: ADAR1, -2, and -3. ADAR1is widely expressed during embryonic and post-natal development and is present as a predom-inantly nuclear, constitutive ADAR1p110 isoformexpressed in all tissues and an additional in-terferon (IFN)–inducible ADAR1p150 isoformthat is found in both the nucleus and the cyto-plasm (5). Adar1−/− (null for both isoforms) andAdar1p150−/− mice die in utero at E11.5 (embry-onic day 11.5) to E12.5, due to failed erythropoi-esis and fetal liver (FL) disintegration (6–8).Only ADAR1 and ADAR2 demonstrate editing

activity in vitro. The AMPA receptor GluA2 pre-mRNA is edited by ADAR2 in the brain, con-verting a glutamine residue to an arginine in afunctionally critical position (9). Adar2−/− micedie from seizures and were rescued by the

genomic substitution of the single edited aden-osine for guanosine, mimicking editing of thetranscript (10). In contrast to this elegant single-substrate paradigm that describes the Adar2−/−

phenotype, no such editing site(s) described todate have resolved the physiological requirementfor ADAR1.To directly determine the contribution of

A-to-I editing to ADAR1’s biological function, wegenerated a constitutive knock-in of an editing-deficient ADAR1 allele in mice (Adar1E861A, whereE861A denotes Glu861→Ala861) (Fig. 1A and fig.S1A), homologous to the human ADAR1E912A

allele, which is catalytically inactive in vitro (11)(also see supplementarymaterials andmethods).In E12.5 whole embryos, ADAR1p110 protein wasdetected in Adar1E861A/E861A samples at compa-rable levels toAdar1+/+ andAdar1E861A/+ controls(Fig. 1B). Additionally, Adar1E861A/E861A embryoshad elevated expression of ADAR1p150 (Fig. 1B).Adar1E861A/+ heterozygous mice were normal,

were present at the expected Mendelian ratio,and had no discernible phenotype. No viableAdar1E861A/E861A pups were born from Adar1E861A/+

intercrosses and were found to be dying at ~E13.5(Fig. 1C). Adar1E861A/E861A yolk sacs were pale,and embryos appeared developmentally delayedcompared with controls (Fig. 1D), despite beingnormal at E12.5 (fig. S1, D and E). At E13.5,Adar1E861A/E861A FL was smaller, with eightfoldfewer viable cells (Fig. 1D and fig. S1E). Weobserved a failure in erythropoiesis with a severeloss of erythroblast populations (Fig. 1, E and F)and increased cell death (Fig. 1G), also apparentin other hematopoietic populations (fig. S2).Consistent with our previous analysis of theADAR1 conditional allele (12), adult hemato-poietic stem cells (HSCs) expressing only thecatalytically inactive ADAR1 could not be main-

tained in vivo and were lost over time (fig. S3).The A-to-I editing activity of ADAR1 is essentialfor embryonic development and themaintenanceof hematopoiesis in vivo.We performed transcriptional profiling on

three independent littermate Adar1+/+ andAdar1E861A/E861A FLs. An absence of ADAR1 edit-ing resulted in the up-regulation of 383 transcripts(log2FC > 2), 258 of which are IFN-stimulatedgenes (ISGs) (Fig. 2A and table S1). Pathway anal-ysis revealed a profound enrichment of signaturesactivated after IFN treatment or viral infection(Fig. 2B and table S1). Of the 50most differentiallyexpressed genes, 44 were up-regulated by eithertype I IFN, type II IFN, or both (Fig. 2C and tableS1). The derepression of ISGs was confirmed byquantitative reverse transcriptase polymerasechain reaction (qRT-PCR) (Fig. 2D). Further-more, we demonstrated that the transcriptionalresponse in Adar1E861A/E861A FLs closely parallelsthat of complete ADAR1 deficiency (12) (fig. S4A).Synthetic dsRNA sequences based on endoge-

nous RNA containing adenosine but not inosinestimulate ISGs and apoptosis in vitro by bindingto MDA5 and RIG-I (13). We defined a gene setassociatedwith the response to IU-dsRNA (inosine-uracil–paired dsRNA) in human cells (13). Genesignatures of bothAdar1E861A/E861A FL andAdar1−/−

HSCs were highly enriched for the IU-dsRNAresponse (Fig. 2E), demonstrating a species-conserved response to dsRNA that is constrainedby the presence of inosine residues. Therefore,the deamination of adenosine in dsRNA byADAR1 is necessary for suppression of the IFNresponse under homeostatic conditions. Fur-thermore, it suggested that the absence of A-to-Isubstitutions in endogenous dsRNAmay initiatethis response.To define ADAR1-specific editing events in vivo,

we analyzed A-to-I (G) mismatches within theRNA sequencing data. Across all samples, 6167A-to-I editing sites were identified: 5540 known(14) and 627 previously undiscovered (Fig. 3A).Strain-specific single-nucleotide polymorphismswere excluded, and only A-to-I mismatch fre-quencies that differed significantly between geno-types were considered (Fig. 3B). Using thesecriteria, 673 A-to-I editing sites were defined(Fig. 3A). Of these, 666 sites had reduced editinginmutants, including hyperedited loci, which hadno detectable editing (Fig. 3B and tables S2 andS3). This confirmed that the Adar1E861A allele iscatalytically inactive and the majority of FL A-to-I editing is ADAR1 dependent. We validated281 of the identified A-to-I sites on independentAdar1E861A/E861A and Adar1+/+ FL samples (15).All tested sites were confirmed as differentiallyedited in both FL and mouse embryonic fibro-blasts (MEFs) (table S4). Sanger sequencingvalidated ADAR1-specific BlcapY2C,Mad2l1,Rbbp4,and Klf1 editing sites from independent FL sam-ples (Fig. 3C).ADAR1 is required for erythropoiesis (Fig. 1,

E to G). Interestingly, 40% (264 of 666) of theADAR1-specific editing sites were within hyper-edited clusters of only three genes—Klf1, Oip5, andOptn—which have peak or restricted expression in

SCIENCE sciencemag.org 4 SEPTEMBER 2015 • VOL 349 ISSUE 6252 1115

1St. Vincent’s Institute of Medical Research, Fitzroy, Victoria3065, Australia. 2Department of Medicine, St. Vincent’sHospital, University of Melbourne, Fitzroy, Victoria 3065,Australia. 3Department of Genetics, Stanford University,Stanford, CA 94305, USA. 4Department of MolecularNeurobiology, Max Planck Institute for Medical Research,69120 Heidelberg, Germany. 5Taconic Biosciences, 51063Cologne, Germany.*These authors contributed equally to this work. †Correspondingauthor. E-mail: [email protected]

RESEARCH | REPORTSon N

ovember 12, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 2: RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of ... · RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself Brian J. Liddicoat,1,2 Robert

the erythroid lineage (table S3). Seventy, 61, and133 ADAR1-specific A-to-I hyperedited loci werewithin long 3′ untranslated regions (3′UTRs) ofKlf1, Oip5, and Optn, respectively. An exampleof hyperediting in Klf1 is depicted in Fig. 3D. Anabsence of ADAR1 editing of these loci providesa possible link to the FL failure in Adar1E861A/E861A

embryos.Modeling of the predicted secondary structure

of the hyperedited 3′UTRs showed that in theabsence of editing there was the potential forlongperfect dsRNAsegments to be formed throughduplexing of repetitive regions (Fig. 3E and fig.S7, A to C). The thermodynamics of inosine basepairing is not defined. Therefore, we modeledsecondary structures for hyperedited substratesin two ways: (i) by predicting secondary struc-tures of Klf1 (Fig. 3E, left), Optn, and Oip5 (fig.S7) 3′UTRs with inosine in place of adenosine,assuming no base pairing, or (ii) by replacingadenosine with guanosine (Fig. 3E, middle), as-

suming wobble base paring. In both cases, thepredicted dsRNA structures have higher free-energy states in the presence of A-to-I or A-to-Gsubstitutions (fig. S7, A to F). Therefore, ex-tensive I-U mismatches would be predicted todestabilize perfect dsRNA stem loops withinhyperedited 3′UTRs (Fig. 3E and fig. S7, A to F).Thus, we hypothesized that ADAR1 editing re-modeled the secondary structure of endogenousRNA to abrogate the formation of long matcheddsRNAs.Thetranscriptionalsignatures intheAdar1E861A/E861A

FL resembled that of RIG-I and MDA5 activa-tion (13). We postulated that in the absence ofADAR1 editing, endogenous dsRNAs—such as the3′UTRs of Klf1, Oip5, and Optn—could be boundby MDA5 and/or RIG-I and could activate thecellular dsRNA response. Short hairpin RNA(shRNA) knockdown of MDA5 rescued prolifer-ation and viability (Fig. 4, A to C) and suppressedISG induction in hematopoietic stem and pro-

genitor cells expressing only the Adar1E861A alleleto control levels, using two independent MDA5shRNAs (fig. S8).Based on the in vitro rescue, we crossed

Adar1E861A/E861A to MDA5−/− (Ifih1−/−) mice. Ini-tially we assessed embryos at E13.5, a timepoint when Adar1E861A/E861A embryos are no longerviable (Fig. 1C). Adar1E861A/E861AIfih1−/− double mu-tant yolk sac, embryo, and FL were comparableto controls and were present at the expectedratio (Fig. 4D and fig. S9A). Erythropoiesis wascompletely rescued, and FL ISGs were equivalentto those of control littermates (Fig. 4, E to G).Most surprisingly, viable Adar1E861A/E861AIfih1−/−

mutants that survived past weaning have beenidentified (fig. S9B). The viable double mutantsare outwardly healthy, albeit slightly smaller thanlittermate controls, and have not demonstratedadditional phenotypes to date (Fig. 4H). The res-cue of both developmental and adult viabilitydemonstrates that MDA5 is the primary sensor

1116 4 SEPTEMBER 2015 • VOL 349 ISSUE 6252 sciencemag.org SCIENCE

Fig. 1. Adar1E861A/E861A embryos die in utero. (A) Schematic of Adar1E861A knock-in allele. (B) ADAR1 protein expression in whole E12.5 embryos of theindicated genotypes. (C) Survival data at the indicated stages. (D) Images of viable E13.5 yolk sacs, embryos, and FL. Scale: yolk sac and embryo, 1 cm;FL, 2 mm. Representative (E) fluorescence-activated cell sorting (FACS) profiles, (F) cell numbers, and (G) frequency of 7AAD+ FL erythroblasts at E13.5.Results are mean ± SEM (+/+, n = 5 embryos; E861A/+, n = 18; E861A/E861A, n = 3). **P < 0.005 and ***P < 0.0005 compared with Adar1+/+. R2 to R5denote erythroblast populations.

RESEARCH | REPORTSon N

ovember 12, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 3: RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of ... · RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself Brian J. Liddicoat,1,2 Robert

of endogenous dsRNA in the absence of ADAR1editing.Unlike ADAR2, the primary role of ADAR1 has

not been clearly defined. Although it was as-sumed that RNA editing was its central function,additional RNA editing–independent roles havebeen proposed. The murine phenotypes and

transcriptional consequences of complete ADAR1deficiency and the specific loss of A-to-I editingare markedly similar, demonstrating that RNAediting is the primary and physiologically mostimportant function of ADAR1. Adar1E861A/E861A

embryos die ~1 to 1.5 days later than Adar1−/−

embryos, suggesting limited contributions from

nonediting functions of ADAR1. We believe thedifference in survival is accounted for by thecatalytically inactive ADAR1 retaining the abilityto sequester immunogenic dsRNA (16). ADAR1sequestration is not sufficient, however, to com-pletely prevent MDA5 recognition of unediteddsRNA and subsequent signaling. Therefore, the

SCIENCE sciencemag.org 4 SEPTEMBER 2015 • VOL 349 ISSUE 6252 1117

Fig. 2. Absence ofediting transcription-ally phenocopies lossof ADAR1. (A) MA plotcomparing geneexpression in WT andE861A E12.5 FL. Reddots, differentiallyexpressed genes;blue dots, differentiallyexpressed ISGs.(B) QuSAGE analysisof the top 100 differen-tial pathway signaturesranked by fold enrich-ment and P value.(C) Heat map of the50 most differentiallyexpressed genes. Blackdots indicate knownISGs. (D) qRT-PCR ofISGs in E861A com-pared with controls inFL and MEFs. Resultsare mean T SEM(n = 3). *P < 0.05,**P < 0.005, and ***P <0.0005 compared withAdar1+/+. (E) IU-dsRNAresponse gene set inE861A compared withWTsamples (left) andAdar1−/− HSCs com-pared with controls(right).

RESEARCH | REPORTSon N

ovember 12, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 4: RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of ... · RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself Brian J. Liddicoat,1,2 Robert

1118 4 SEPTEMBER 2015 • VOL 349 ISSUE 6252 sciencemag.org SCIENCE

Fig. 3. Defining the ADAR1 FL editome. (A) Summary of A-to-I editing siteanalysis in FL. SNPs, single-nucleotide polymorphisms; ANOVA, analysis ofvariance; ncRNA, noncoding RNA. (B) Mean editing difference for sites with>20 reads (left, n = 1634). Mean editing frequency of differentially edited sitesbetweenE861A and controls (right, n=673). (C) GenomicDNA (gDNA) (bottom)and complementary DNA (cDNA) (top andmiddle) Sanger sequencing validation

of editing sites. Red arrows highlight edited adenosine. (D) Integrative GenomicsViewer image of Klf1 in WT E12.5 FL and predicted secondary structure of 3′UTR.The red line denotes the region depicted in (E). (E) Predicted secondary structureofa212–basepairdsRNAstemloop fromthe3′UTRofKlf1,with inosine (IU-dsRNA,left) and guanosine (GU-dsRNA, middle) in place of adenosine at the editedsites. Right, predicted secondary structure with no A-to-I editing.

RESEARCH | REPORTSon N

ovember 12, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 5: RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of ... · RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself Brian J. Liddicoat,1,2 Robert

extended survival of the editing-deficient animalsreflects a delay rather than a fundamental differ-ence in the presentation of the same phenotype.The data from the Adar1E861A mutants are

consistent with the type I interferonopathiesof Aicardi-Goutières syndrome (AGS) patientsbearing ADAR1 mutations (17, 18). Editing ofendogenous transcripts in ADAR1-mutant AGSpatients is probably reduced, leading to retentionof paired endogenous dsRNA that can be sensedby MDA5. Consistent with this, gain-of-functionmutations ofMDA5 have been identified in non–ADAR1-mutant AGS patients (18). AGS MDA5mutations caused higher-affinity binding to self-dsRNA, resulting in the inappropriate detection

of self-dsRNA (18). Thus, the physiological func-tion of ADAR1 is probably conserved inmammals.Approximately half of the mammalian genome

is composed of noncoding retrotransposons suchas SINEs and Alus (19, 20), which typically formdsRNA duplexes. Retrotransposons are subjectedto extensive A-to-I RNAediting (3, 4). The locationof repetitive elementsmay determine their immu-nogenicity. Retrotransposons located within in-trons do not persist in the cytosol and thereforecannot activate MDA5. Repetitive elements in3′UTRs, though rare (4), can be retained andform duplexes, harboring the potential for recog-nition by MDA5. We propose that hypereditingof self-dsRNA by ADAR1 (such as the 3′UTRs

of Klf1, Optn, and Oip5) generates multiple I-Umismatches that act to prevent MDA5 oligomer-ization (21). In the absence of ADAR1 editing,long dsRNA stem loops can form that activateMDA5 (fig. S10). However, we cannot rule out analternate possibility that edited substrates pref-erentially bind MDA5 to prevent its activationby other (unidentified and nonedited) dsRNAtranscripts (13).Concurrent ablation of MAVS, the downstream

adaptor of MDA5 and RIG-I, rescues ADAR1-nullmice to birth (22), demonstrating a role forADAR1 in the suppression of the RLR pathway.Our study specifically identifies the critical cyto-solic sensor upstream of MAVS and demonstrates

SCIENCE sciencemag.org 4 SEPTEMBER 2015 • VOL 349 ISSUE 6252 1119

Fig. 4. Loss ofMDA5 rescuesAdar1E861A/E861Aviability. (A) LKS+ cells isolatedfromRosa26CreERT2Adar1fl/+ (D/+) andRosa26CreERT2Adar1fl/E861A (D/E861A)infected with pLKO.1 empty vector, shGFP, or (B) two independent shMDA5[shMDA5(1) and shMDA5(2)] were cultured for 8 days. Results aremean T SEM(n = 3). **P < 0.005 and ***P < 0.0005 compared with D/+. n.s, not signifi-cant. (C) Analysis of apoptosis on day 8. (D) Images of viable E13.5 yolk sac,

embryo, and FL of the indicated genotype (all Ifih1−/−). Scale: yolk sac and em-bryo, 5mm; FL, 1.6mm.Representative (E) FACSprofiles and (F) numbers of FLerythrocytes at E13.5. (G) E13.5 FL qRT-PCR of ISGs. Results are mean T SEM(+/+, n = 2; E861A/+, n = 8; E861A/E861A, n = 4). *P < 0.05 compared withAdar1+/+Ifih1−/− controls. (H) Photo of an Adar1E861A/E861A Ifih1−/− mouse andan Adar1E861A/+Ifih1+/− littermate at 26 days of age.

RESEARCH | REPORTSon N

ovember 12, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 6: RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of ... · RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself Brian J. Liddicoat,1,2 Robert

that the Adar1E861A/E861A phenotype and, by ex-tension, Adar1−/− can be ascribed to the lack ofediting of multiple substrates, resulting in theinappropriate activation of MDA5. We speculatethat these unedited transcripts are sensed asnonself by MDA5 and activate innate immunesignaling. ADAR1’s primary physiological functionis to edit endogenous dsRNA to prevent sensingof endogenous dsRNA as nonself by MDA5.

REFERENCES AND NOTES

1. P. Danecek et al., Genome Biol. 13, R26 (2012).2. J. B. Li et al., Science 324, 1210–1213 (2009).3. G. Ramaswami et al., Nat. Methods 9, 579–581 (2012).4. Y. Neeman, E. Y. Levanon, M. F. Jantsch, E. Eisenberg, RNA 12,

1802–1809 (2006).5. J. B. Patterson, C. E. Samuel, Mol. Cell. Biol. 15, 5376–5388

(1995).6. J. C. Hartner et al., J. Biol. Chem. 279, 4894–4902 (2004).7. Q. Wang et al., J. Biol. Chem. 279, 4952–4961 (2004).8. S. V. Ward et al., Proc. Natl. Acad. Sci. U.S.A. 108, 331–336

(2011).9. M. Higuchi et al., Cell 75, 1361–1370 (1993).10. M. Higuchi et al., Nature 406, 78–81 (2000).11. F. Lai, R. Drakas, K. Nishikura, J. Biol. Chem. 270, 17098–17105

(1995).12. J. C. Hartner, C. R. Walkley, J. Lu, S. H. Orkin, Nat. Immunol. 10,

109–115 (2009).13. P. Vitali, A. D. Scadden, Nat. Struct. Mol. Biol. 17, 1043–1050

(2010).14. G. Ramaswami, J. B. Li, Nucleic Acids Res. 42, D109–D113

(2014).15. R. Zhang et al., Nat. Methods 11, 51–54 (2014).16. M. C. Washburn et al., Cell Reports 6, 599–607 (2014).17. G. I. Rice et al., Nat. Genet. 44, 1243–1248 (2012).18. G. I. Rice et al., Nat. Genet. 46, 503–509 (2014).19. E. S. Lander et al., Nature 409, 860–921 (2001).20. Mouse Genome Sequencing Consortium, Nature 420, 520–562

(2002).21. B. Wu et al., Cell 152, 276–289 (2013).22. N. M. Mannion et al., Cell Reports 9, 1482–1494

(2014).

ACKNOWLEDGMENTS

We thank V. Sankaran, S. Orkin, L. Purton, and J. Heierhorst fordiscussion and SVH BioResources Centre for animal care. Datasets described in the paper are deposited in Gene ExpressionOmnibus (accession number GSE58917). This work was supportedby the Leukaemia Foundation (C.R.W.), a Leukaemia FoundationPh.D. scholarship (B.J.L), National Health and Medical ResearchCouncil (NHMRC) Project Grant 1021216, NHMRC CareerDevelopment Award 559016 (C.R.W.), a German AcademicExchange Service Postdoctoral Fellowship (R.P.), a StanfordUniversity Dean’s Fellowship (R.P.), Stanford Genome TrainingProgram (NIH grant T32 HG000044) and Stanford GraduateFellowship (G.R.), NIH grant R01GM102484 (J.B.L.), the EllisonMedical Foundation (J.B.L.), and the Stanford UniversityDepartment of Genetics (J.B.L.). This work was also supported inpart by the Victorian State Government Operational InfrastructureSupport Scheme (to St. Vincent’s Institute of Medical Research). C.R.W. was the Leukaemia Foundation Phillip Desbrow SeniorResearch Fellow. J.C.H. is an employee of Taconic Biosciences.Taconic Biosciences had no role in the preparation and content ofthis Report. All other authors declare no conflicts of interest.Author contributions: M.H., P.H.S., and J.C.H. generated the knock-in mouse line; B.J.L., R.P., A.M.C., G.R., J.C.H., J.B.L., and C.R.W.performed experiments and analyzed and interpreted data; M.H.,P.H.S., and J.C.H. provided intellectual input and conceptualadvice; and B.J.L. and C.R.W. wrote the manuscript.

SUPPLEMENTARY MATERIALS

www.sciencemag.org/content/349/6252/1115/suppl/DC1Materials and MethodsSupplementary TextFigs. S1 to S10Tables S1 to S4References (23–41)

1 June 2015; accepted 13 July 2015Published online 23 July 201510.1126/science.aac7049

DNA SEGREGATION

Structures of archaeal DNAsegregation machinery revealbacterial and eukaryotic linkagesMaria A. Schumacher,1* Nam K. Tonthat,1 Jeehyun Lee,1

Fernando A. Rodriguez-Castañeda,2 Naga Babu Chinnam,1 Anne K. Kalliomaa-Sanford,2

Irene W. Ng,2 Madhuri T. Barge,2 Porsha L. R. Shaw,1 Daniela Barillà2*

Although recent studies have provided a wealth of information about archaeal biology,nothing is known about the molecular basis of DNA segregation in these organisms. Here,we unveil the machinery and assembly mechanism of the archaeal Sulfolobus pNOB8partition system. This system uses three proteins: ParA; an atypical ParB adaptor; and acentromere-binding component, AspA. AspA utilizes a spreading mechanism to create aDNA superhelix onto which ParB assembles. This supercomplex links to the ParA motor,which contains a bacteria-like Walker motif. The C domain of ParB harbors structuralsimilarity to CenpA, which dictates eukaryotic segregation. Thus, this archaeal systemcombines bacteria-like and eukarya-like components, which suggests the possibleconservation of DNA segregation principles across the three domains of life.

DNA segregation or partition is an essentialbiological process ensuring faithful ge-nomic transmission. The best-understoodsegregation systems at the molecular levelare those used by bacterial plasmids.

These simplified systems consist of a DNA cen-tromere; ParA nucleoside triphosphatase (NTPase);and centromere-binding protein (CBP), ParB (1–6).The most common partition apparatuses useWalker-box NTPases (7). The segregation sys-tems that have been identified on bacterial chro-mosomes also harbor Walker NTPases, althoughtheir mechanisms are less clear (1–6). In con-trast to bacterial partition, eukaryotic partitionis highly complex. However, the linchpin in eu-karyotic segregation is the histone protein, CenpA,which is deposited in place of histoneH3 at DNAcentromeres and dictates assembly of the segre-gation machinery (8–11). Although some pro-gress has been made in understanding DNAsegregation in eukarya and bacteria, virtuallynothing is known about the molecular processof segregation in archaea, the third domain oflife (12–14).To gain insight into the underpinnings of

archaeal segregation, we performed a moleculardissection of the proteins encoded on the plas-mid pNOB8 partition cassette harbored in Sulfo-lobus NOB8H2 (15). This cassette contains threeopen reading frames (ORFs): orf44, orf45, andorf46. orf46 and orf45 encode 315- and 470-residue proteins, respectively, which show 33 to37% and 42 to 58% sequence similarity to bac-terial ParA and ParB proteins. orf44 generatesa 93-residue protein that shows no sequence

similarity to any characterized partition protein.The organization of the orf44-parB-parA genes(fig. S1A) is reminiscent of bacterial partitioncassettes; however, the cluster is tricistronic, un-like typical bicistronic bacterial systems (1–4).Hypothesizing that the pNOB8 centromeremay be located either 5′ or 3′ of the cassette asin bacteria, we analyzed pNOB8 protein bindingto these regions. Unlike its bacterial counterparts,pNOB8 ParB only bound DNA nonspecifically(fig. S1B). Orf44, however, bound the upstreamDNAwith high affinity [apparent dissociationconstant (Kdapp) = ~50 nM] (Fig. 1A). Therefore,we named this CBP, archaeal segregation pro-tein A (AspA). Deoxyribonuclease I (DNase I)footprinting showed that AspA interacted witha 23–base pair (bp) palindrome in the upstreamregion and that increasing AspA concentrationsled to spreading around this site (Fig. 1B). Hence,like bacterial ParB proteins, AspA spreads non-specifically to DNA adjacent to its centromereto formanextended “partition-complex.”Althoughpoorly characterized, higher-order partition-complexes are central to segregation, as theymediate stabilizing, dynamic interactions withParA assemblages (1–4). Indeed, the CBP-NTPaseinteraction is key to the partition process. Bio-chemical experiments, however, showed thatAspA does not bind pNOB8 ParA. Rather, ParBbound both AspA [dissociation constant (Kd =12 mM)] and ParA (Kd = 17 mM), which indicatedthat it acts as an adaptor (fig. S1C). Thus, thepNOB8 partition system is composed of theCBP AspA, adaptor ParB, and ParA NTPase.This finding prompted us to perform BLASTsearches for the occurrence of aspA-parB-parAcassettes on archaeal genomes. The results re-vealed that this tripartite cluster of genes is wide-spread across different crenarchaeal genera and isharbored on both chromosomes and plasmids(fig. S2).

1120 4 SEPTEMBER 2015 • VOL 349 ISSUE 6252 sciencemag.org SCIENCE

1Department of Biochemistry, Duke University School ofMedicine, 243 Nanaline H. Duke, Box 3711, Durham, NC27710, USA. 2Department of Biology, University of York, YorkYO10 5DD, UK.*Corresponding author. E-mail: [email protected](M.A.S.); [email protected] (D.B.)

RESEARCH | REPORTSon N

ovember 12, 2020

http://science.sciencem

ag.org/D

ownloaded from

Page 7: RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of ... · RNA EDITING RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself Brian J. Liddicoat,1,2 Robert

RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself

Seeburg and Carl R. WalkleyBrian J. Liddicoat, Robert Piskol, Alistair M. Chalk, Gokul Ramaswami, Miyoko Higuchi, Jochen C. Hartner, Jin Billy Li, Peter H.

originally published online July 23, 2015DOI: 10.1126/science.aac7049 (6252), 1115-1120.349Science 

, this issue p. 1115Scienceresponse to dsRNA in the cytoplasm, because dsRNA is also a marker of viral infection.portions of cellular mRNAs. ADAR1-directed editing of these cellular targets is critical to avoid activation of an immunein vivo targets of the principal editing enzyme, ADAR1, are long double-stranded RNA (dsRNA) structures in noncoding

show that theet al.This RNA ''editing'' is mediated by adenosine deaminase acting on RNA (ADAR) enzymes. Liddicoat Adenosine bases in messenger RNA (mRNAs) can be enzymatically modified and changed into inosine bases.

RNA editing helps identify cellular RNAs

ARTICLE TOOLS http://science.sciencemag.org/content/349/6252/1115

MATERIALSSUPPLEMENTARY http://science.sciencemag.org/content/suppl/2015/07/22/science.aac7049.DC1

REFERENCES

http://science.sciencemag.org/content/349/6252/1115#BIBLThis article cites 41 articles, 11 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.ScienceScience, 1200 New York Avenue NW, Washington, DC 20005. The title (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement ofScience

Copyright © 2015, American Association for the Advancement of Science

on Novem

ber 12, 2020

http://science.sciencemag.org/

Dow

nloaded from