Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.127
Filter
Add more filters

Publication year range
1.
Cell ; 186(5): 999-1012.e20, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36764292

ABSTRACT

Adenosine-to-inosine RNA editing has been proposed to be involved in a bacterial anti-phage defense system called RADAR. RADAR contains an adenosine triphosphatase (RdrA) and an adenosine deaminase (RdrB). Here, we report cryo-EM structures of RdrA, RdrB, and currently identified RdrA-RdrB complexes in the presence or absence of RNA and ATP. RdrB assembles into a dodecameric cage with catalytic pockets facing outward, while RdrA adopts both autoinhibited tetradecameric and activation-competent heptameric rings. Structural and functional data suggest a model in which RNA is loaded through the bottom section of the RdrA ring and translocated along its inner channel, a process likely coupled with ATP-binding status. Intriguingly, up to twelve RdrA rings can dock one RdrB cage with precise alignments between deaminase catalytic pockets and RNA-translocation channels, indicative of enzymatic coupling of RNA translocation and deamination. Our data uncover an interesting mechanism of enzymatic coupling and anti-phage defense through supramolecular assemblies.


Subject(s)
Adenosine Triphosphate , RNA , Adenosine Deaminase/genetics
2.
Annu Rev Biochem ; 88: 191-220, 2019 06 20.
Article in English | MEDLINE | ID: mdl-30883196

ABSTRACT

Programmable nucleases and deaminases, which include zinc-finger nucleases, transcription activator-like effector nucleases, CRISPR RNA-guided nucleases, and RNA-guided base editors, are now widely employed for the targeted modification of genomes in cells and organisms. These gene-editing tools hold tremendous promise for therapeutic applications. Importantly, these nucleases and deaminases may display off-target activity through the recognition of near-cognate DNA sequences to their target sites, resulting in collateral damage to the genome in the form of local mutagenesis or genomic rearrangements. For therapeutic genome-editing applications with these classes of programmable enzymes, it is essential to measure and limit genome-wide off-target activity. Herein, we discuss the key determinants of off-target activity for these systems. We describe various cell-based and cell-free methods for identifying genome-wide off-target sites and diverse strategies that have been developed for reducing the off-target activity of programmable gene-editing enzymes.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing/methods , Protein Engineering/methods , RNA, Guide, Kinetoplastida/genetics , APOBEC Deaminases/genetics , APOBEC Deaminases/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Artifacts , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Protein 9/metabolism , Endonucleases/genetics , Endonucleases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Transfer Techniques , Genome, Human , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , RNA, Guide, Kinetoplastida/metabolism , Software
3.
Cell ; 172(4): 640-642, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29425484

ABSTRACT

Discrimination between viral and self-derived nucleic acid species is crucial in maintaining effective antiviral immunity whilst avoiding autoinflammation. Ahmad et al. and Chung et al. delineate the consequences of MDA5 gain of function and loss of ADAR1 activity, highlighting the blurring of the concept of self and non-self when considering endogenous retroelements.


Subject(s)
RNA Editing , RNA , Adenosine Deaminase/genetics , DEAD-box RNA Helicases/genetics , Humans , Inflammation , RNA-Binding Proteins , Self Tolerance
4.
Cell ; 172(4): 811-824.e14, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29395325

ABSTRACT

Type I interferon (IFN) is produced when host sensors detect foreign nucleic acids, but how sensors differentiate self from nonself nucleic acids, such as double-stranded RNA (dsRNA), is incompletely understood. Mutations in ADAR1, an adenosine-to-inosine editing enzyme of dsRNA, cause Aicardi-Goutières syndrome, an autoinflammatory disorder associated with spontaneous interferon production and neurologic sequelae. We generated ADAR1 knockout human cells to explore ADAR1 substrates and function. ADAR1 primarily edited Alu elements in RNA polymerase II (pol II)-transcribed mRNAs, but not putative pol III-transcribed Alus. During the IFN response, ADAR1 blocked translational shutdown by inhibiting hyperactivation of PKR, a dsRNA sensor. ADAR1 dsRNA binding and catalytic activities were required to fully prevent endogenous RNA from activating PKR. Remarkably, ADAR1 knockout neuronal progenitor cells exhibited MDA5 (dsRNA sensor)-dependent spontaneous interferon production, PKR activation, and cell death. Thus, human ADAR1 regulates sensing of self versus nonself RNA, allowing pathogen detection while avoiding autoinflammation.


Subject(s)
Adenosine Deaminase/metabolism , Alu Elements , Autoimmune Diseases of the Nervous System/metabolism , Nervous System Malformations/metabolism , Neural Stem Cells/metabolism , Protein Biosynthesis , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/immunology , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Cell Death/genetics , Cell Death/immunology , Gene Knockout Techniques , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/immunology , Interferon-Induced Helicase, IFIH1/metabolism , Nervous System Malformations/genetics , Nervous System Malformations/immunology , Neural Stem Cells/cytology , Neural Stem Cells/immunology , Neural Stem Cells/pathology , RNA Polymerase II/genetics , RNA Polymerase II/immunology , RNA Polymerase II/metabolism , RNA, Double-Stranded/genetics , RNA, Double-Stranded/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology , eIF-2 Kinase/metabolism
5.
Immunity ; 56(5): 979-997.e11, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37100060

ABSTRACT

Immune cell trafficking constitutes a fundamental component of immunological response to tissue injury, but the contribution of intrinsic RNA nucleotide modifications to this response remains elusive. We report that RNA editor ADAR2 exerts a tissue- and stress-specific regulation of endothelial responses to interleukin-6 (IL-6), which tightly controls leukocyte trafficking in IL-6-inflamed and ischemic tissues. Genetic ablation of ADAR2 from vascular endothelial cells diminished myeloid cell rolling and adhesion on vascular walls and reduced immune cell infiltration within ischemic tissues. ADAR2 was required in the endothelium for the expression of the IL-6 receptor subunit, IL-6 signal transducer (IL6ST; gp130), and subsequently, for IL-6 trans-signaling responses. ADAR2-induced adenosine-to-inosine RNA editing suppressed the Drosha-dependent primary microRNA processing, thereby overwriting the default endothelial transcriptional program to safeguard gp130 expression. This work demonstrates a role for ADAR2 epitranscriptional activity as a checkpoint in IL-6 trans-signaling and immune cell trafficking to sites of tissue injury.


Subject(s)
Interleukin-6 , RNA , Endothelial Cells/metabolism , Cytokine Receptor gp130 , Endothelium/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism
6.
Mol Cell ; 83(21): 3760-3762, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37922869

ABSTRACT

In this issue, Hu and Heraud-Farlow et al.1 demonstrate that ADAR1 dsRNA editing and dsRNA binding activities are critical to repress MDA5 and PKR, respectively, and that PKR and MDA5 act in concert to induce fatality in ADAR1 KO mice.


Subject(s)
Adenosine Deaminase , RNA, Double-Stranded , Mice , Animals , RNA, Double-Stranded/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism
7.
Mol Cell ; 83(21): 3869-3884.e7, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37797622

ABSTRACT

Effective immunity requires the innate immune system to distinguish foreign nucleic acids from cellular ones. Cellular double-stranded RNAs (dsRNAs) are edited by the RNA-editing enzyme ADAR1 to evade being recognized as viral dsRNA by cytoplasmic dsRNA sensors, including MDA5 and PKR. The loss of ADAR1-mediated RNA editing of cellular dsRNA activates MDA5. Additional RNA-editing-independent functions of ADAR1 have been proposed, but a specific mechanism has not been delineated. We now demonstrate that the loss of ADAR1-mediated RNA editing specifically activates MDA5, whereas loss of the cytoplasmic ADAR1p150 isoform or its dsRNA-binding activity enabled PKR activation. Deleting both MDA5 and PKR resulted in complete rescue of the embryonic lethality of Adar1p150-/- mice to adulthood, contrasting with the limited or no rescue by removing MDA5 or PKR alone. Our findings demonstrate that MDA5 and PKR are the primary in vivo effectors of fatal autoinflammation following the loss of ADAR1p150.


Subject(s)
Immunity, Innate , RNA, Double-Stranded , Animals , Mice , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Cytoplasm/metabolism , Immunity, Innate/genetics , RNA, Double-Stranded/genetics
8.
Immunity ; 54(9): 1909-1911, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525334

ABSTRACT

Some RNAs can assume a Z conformation, an unusual, left-handed turn. In this issue of Immunity, three studies report that mutations in the Zα-RNA binding domain of the adenosine deaminase ADAR1 are sufficient to induce autoinflammatory disease in mice, which models human Aicardí-Goutières syndrome, highlighting the important role of Z-RNA editing in limiting innate immune recognition of endogenous RNA.


Subject(s)
Autoimmune Diseases of the Nervous System , RNA , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Mice , RNA/genetics , RNA Editing , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
9.
Immunity ; 54(9): 1976-1988.e7, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525338

ABSTRACT

Mutations in the adenosine-to-inosine RNA-editing enzyme ADAR1 p150, including point mutations in the Z-RNA recognition domain Zα, are associated with Aicardi-Goutières syndrome (AGS). Here, we examined the in vivo relevance of ADAR1 binding of Z-RNA. Mutation of W197 in Zα, which abolished Z-RNA binding, reduced RNA editing. Adar1W197A/W197A mice displayed severe growth retardation after birth, broad expression of interferon-stimulated genes (ISGs), and abnormal development of multiple organs. Notably, malformation of the brain was accompanied by white matter vacuolation and gliosis, reminiscent of AGS-associated encephalopathy. Concurrent deletion of the double-stranded RNA sensor MDA5 ameliorated these abnormalities. ADAR1 (W197A) expression increased in a feedback manner downstream of type I interferons, resulting in increased RNA editing at a subset of, but not all, ADAR1 target sites. This increased expression did not ameliorate inflammation in Adar1W197A/W197A mice. Thus, editing of select endogenous RNAs by ADAR1 is essential for preventing inappropriate MDA5-mediated inflammation, with relevance to the pathogenesis of AGS.


Subject(s)
Adenosine Deaminase/genetics , Autoimmune Diseases of the Nervous System/genetics , Nervous System Malformations/genetics , RNA Editing/genetics , RNA, Double-Stranded/genetics , Adenosine Deaminase/metabolism , Animals , Autoimmune Diseases of the Nervous System/physiopathology , Disease Models, Animal , Interferon-Induced Helicase, IFIH1/metabolism , Mice , Mutation , Nervous System Malformations/physiopathology , RNA, Double-Stranded/metabolism
10.
Immunity ; 54(9): 1961-1975.e5, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525337

ABSTRACT

Nucleic acids are powerful triggers of innate immunity and can adopt the Z-conformation, an unusual left-handed double helix. Here, we studied the biological function(s) of Z-RNA recognition by the adenosine deaminase ADAR1, mutations in which cause Aicardi-Goutières syndrome. Adar1mZα/mZα mice, bearing two point mutations in the Z-nucleic acid binding (Zα) domain that abolish Z-RNA binding, displayed spontaneous induction of type I interferons (IFNs) in multiple organs, including in the lung, where both stromal and hematopoietic cells showed IFN-stimulated gene (ISG) induction. Lung neutrophils expressed ISGs induced by the transcription factor IRF3, indicating an initiating role for neutrophils in this IFN response. The IFN response in Adar1mZα/mZα mice required the adaptor MAVS, implicating cytosolic RNA sensing. Adenosine-to-inosine changes were enriched in transposable elements and revealed a specific requirement of ADAR1's Zα domain in editing of a subset of RNAs. Thus, endogenous RNAs in Z-conformation have immunostimulatory potential curtailed by ADAR1, with relevance to autoinflammatory disease in humans.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Adenosine Deaminase/genetics , Interferon Type I/immunology , RNA, Double-Stranded/genetics , Adenosine/genetics , Adenosine/metabolism , Animals , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , Inosine/genetics , Inosine/metabolism , Interferon Type I/genetics , Mice , Mutation , Nervous System Malformations/genetics , Nervous System Malformations/immunology , RNA Editing/genetics , RNA, Double-Stranded/metabolism
11.
12.
Mol Cell ; 81(11): 2374-2387.e3, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33905683

ABSTRACT

Adenosine-to-inosine editing is catalyzed by ADAR1 at thousands of sites transcriptome-wide. Despite intense interest in ADAR1 from physiological, bioengineering, and therapeutic perspectives, the rules of ADAR1 substrate selection are poorly understood. Here, we used large-scale systematic probing of ∼2,000 synthetic constructs to explore the structure and sequence context determining editability. We uncover two structural layers determining the formation and propagation of A-to-I editing, independent of sequence. First, editing is robustly induced at fixed intervals of 35 bp upstream and 30 bp downstream of structural disruptions. Second, editing is symmetrically introduced on opposite sites on a double-stranded structure. Our findings suggest a recursive model for RNA editing, whereby the structural alteration induced by the editing at one site iteratively gives rise to the formation of an additional editing site at a fixed periodicity, serving as a basis for the propagation of editing along and across both strands of double-stranded RNA structures.


Subject(s)
Adenosine Deaminase/genetics , Adenosine/metabolism , Inosine/metabolism , RNA Editing , RNA, Double-Stranded/genetics , RNA-Binding Proteins/genetics , A549 Cells , Adenosine/genetics , Adenosine Deaminase/metabolism , Animals , Base Pairing , HEK293 Cells , Humans , Inosine/genetics , MCF-7 Cells , Mice , NIH 3T3 Cells , Nucleic Acid Conformation , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/metabolism
13.
Nat Rev Mol Cell Biol ; 17(2): 83-96, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26648264

ABSTRACT

Adenosine deaminases acting on RNA (ADARs) convert adenosine to inosine in double-stranded RNA. This A-to-I editing occurs not only in protein-coding regions of mRNAs, but also frequently in non-coding regions that contain inverted Alu repeats. Editing of coding sequences can result in the expression of functionally altered proteins that are not encoded in the genome, whereas the significance of Alu editing remains largely unknown. Certain microRNA (miRNA) precursors are also edited, leading to reduced expression or altered function of mature miRNAs. Conversely, recent studies indicate that ADAR1 forms a complex with Dicer to promote miRNA processing, revealing a new function of ADAR1 in the regulation of RNA interference.


Subject(s)
Adenosine Deaminase/genetics , Adenosine/metabolism , Genome , Inosine/metabolism , RNA Editing , RNA, Messenger/genetics , Adenosine Deaminase/metabolism , Alu Elements , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Signal Transduction
14.
Cell ; 153(3): 575-89, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23622242

ABSTRACT

Adenosine deaminases acting on RNA (ADARs) are involved in RNA editing that converts adenosine residues to inosine specifically in double-stranded RNAs. In this study, we investigated the interaction of the RNA editing mechanism with the RNA interference (RNAi) machinery and found that ADAR1 forms a complex with Dicer through direct protein-protein interaction. Most importantly, ADAR1 increases the maximum rate (Vmax) of pre-microRNA (miRNA) cleavage by Dicer and facilitates loading of miRNA onto RNA-induced silencing complexes, identifying a new role of ADAR1 in miRNA processing and RNAi mechanisms. ADAR1 differentiates its functions in RNA editing and RNAi by the formation of either ADAR1/ADAR1 homodimer or Dicer/ADAR1 heterodimer complexes, respectively. As expected, the expression of miRNAs is globally inhibited in ADAR1(-/-) mouse embryos, which, in turn, alters the expression of their target genes and might contribute to their embryonic lethal phenotype.


Subject(s)
Adenosine Deaminase/metabolism , DEAD-box RNA Helicases/metabolism , RNA Interference , RNA Processing, Post-Transcriptional , Ribonuclease III/metabolism , Adenosine Deaminase/chemistry , Adenosine Deaminase/genetics , Animals , Base Sequence , DEAD-box RNA Helicases/chemistry , Dimerization , Embryo, Mammalian/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , MicroRNAs/metabolism , Molecular Sequence Data , Protein Interaction Domains and Motifs , RNA, Small Interfering/metabolism , RNA-Binding Proteins , Ribonuclease III/chemistry , Up-Regulation
15.
Nature ; 607(7920): 776-783, 2022 07.
Article in English | MEDLINE | ID: mdl-35859176

ABSTRACT

Mutations of the ADAR1 gene encoding an RNA deaminase cause severe diseases associated with chronic activation of type I interferon (IFN) responses, including Aicardi-Goutières syndrome and bilateral striatal necrosis1-3. The IFN-inducible p150 isoform of ADAR1 contains a Zα domain that recognizes RNA with an alternative left-handed double-helix structure, termed Z-RNA4,5. Hemizygous ADAR1 mutations in the Zα domain cause type I IFN-mediated pathologies in humans2,3 and mice6-8; however, it remains unclear how the interaction of ADAR1 with Z-RNA prevents IFN activation. Here we show that Z-DNA-binding protein 1 (ZBP1), the only other protein in mammals known to harbour Zα domains9, promotes type I IFN activation and fatal pathology in mice with impaired ADAR1 function. ZBP1 deficiency or mutation of its Zα domains reduced the expression of IFN-stimulated genes and largely prevented early postnatal lethality in mice with hemizygous expression of ADAR1 with mutated Zα domain (Adar1mZα/- mice). Adar1mZα/- mice showed upregulation and impaired editing of endogenous retroelement-derived complementary RNA reads, which represent a likely source of Z-RNAs activating ZBP1. Notably, ZBP1 promoted IFN activation and severe pathology in Adar1mZα/- mice in a manner independent of RIPK1, RIPK3, MLKL-mediated necroptosis and caspase-8-dependent apoptosis, suggesting a novel mechanism of action. Thus, ADAR1 prevents endogenous Z-RNA-dependent activation of pathogenic type I IFN responses by ZBP1, suggesting that ZBP1 could contribute to type I interferonopathies caused by ADAR1 mutations.


Subject(s)
Adenosine Deaminase , Interferon Type I , RNA-Binding Proteins , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Apoptosis , Caspase 8/metabolism , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Mice , Mutation , Necroptosis , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
16.
Nature ; 608(7923): 569-577, 2022 08.
Article in English | MEDLINE | ID: mdl-35922514

ABSTRACT

A major challenge in human genetics is to identify the molecular mechanisms of trait-associated and disease-associated variants. To achieve this, quantitative trait locus (QTL) mapping of genetic variants with intermediate molecular phenotypes such as gene expression and splicing have been widely adopted1,2. However, despite successes, the molecular basis for a considerable fraction of trait-associated and disease-associated variants remains unclear3,4. Here we show that ADAR-mediated adenosine-to-inosine RNA editing, a post-transcriptional event vital for suppressing cellular double-stranded RNA (dsRNA)-mediated innate immune interferon responses5-11, is an important potential mechanism underlying genetic variants associated with common inflammatory diseases. We identified and characterized 30,319 cis-RNA editing QTLs (edQTLs) across 49 human tissues. These edQTLs were significantly enriched in genome-wide association study signals for autoimmune and immune-mediated diseases. Colocalization analysis of edQTLs with disease risk loci further pinpointed key, putatively immunogenic dsRNAs formed by expected inverted repeat Alu elements as well as unexpected, highly over-represented cis-natural antisense transcripts. Furthermore, inflammatory disease risk variants, in aggregate, were associated with reduced editing of nearby dsRNAs and induced interferon responses in inflammatory diseases. This unique directional effect agrees with the established mechanism that lack of RNA editing by ADAR1 leads to the specific activation of the dsRNA sensor MDA5 and subsequent interferon responses and inflammation7-9. Our findings implicate cellular dsRNA editing and sensing as a previously underappreciated mechanism of common inflammatory diseases.


Subject(s)
Adenosine Deaminase , Genetic Predisposition to Disease , Immune System Diseases , Inflammation , RNA Editing , RNA, Double-Stranded , Adenosine/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Alu Elements/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Genome-Wide Association Study , Humans , Immune System Diseases/genetics , Immune System Diseases/immunology , Immune System Diseases/pathology , Immunity, Innate , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inosine/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Interferons/genetics , Interferons/immunology , Quantitative Trait Loci/genetics , RNA Editing/genetics , RNA, Double-Stranded/genetics , RNA-Binding Proteins/metabolism
17.
Nature ; 607(7920): 769-775, 2022 07.
Article in English | MEDLINE | ID: mdl-35859177

ABSTRACT

The RNA-editing enzyme ADAR1 is essential for the suppression of innate immune activation and pathology caused by aberrant recognition of self-RNA, a role it carries out by disrupting the duplex structure of endogenous double-stranded RNA species1,2. A point mutation in the sequence encoding the Z-DNA-binding domain (ZBD) of ADAR1 is associated with severe autoinflammatory disease3-5. ZBP1 is the only other ZBD-containing mammalian protein6, and its activation can trigger both cell death and transcriptional responses through the kinases RIPK1 and RIPK3, and the protease caspase 8 (refs. 7-9). Here we show that the pathology caused by alteration of the ZBD of ADAR1 is driven by activation of ZBP1. We found that ablation of ZBP1 fully rescued the overt pathology caused by ADAR1 alteration, without fully reversing the underlying inflammatory program caused by this alteration. Whereas loss of RIPK3 partially phenocopied the protective effects of ZBP1 ablation, combined deletion of caspase 8 and RIPK3, or of caspase 8 and MLKL, unexpectedly exacerbated the pathogenic effects of ADAR1 alteration. These findings indicate that ADAR1 is a negative regulator of sterile ZBP1 activation, and that ZBP1-dependent signalling underlies the autoinflammatory pathology caused by alteration of ADAR1.


Subject(s)
Adenosine Deaminase , Immune System Diseases , Inflammation , Mutation , RNA-Binding Proteins , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Animals , Caspase 8/genetics , Caspase 8/metabolism , Cell Death , Gene Deletion , Immune System Diseases/genetics , Immune System Diseases/metabolism , Immune System Diseases/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mammals/genetics , Protein Kinases/deficiency , Protein Kinases/genetics , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction
18.
Immunol Rev ; 322(1): 148-156, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38033164

ABSTRACT

Severe combined immune deficiency due to adenosine deaminase deficiency (ADA SCID) is an inborn error of immunity with pan-lymphopenia, due to accumulated cytotoxic adenine metabolites. ADA SCID has been treated using gene therapy with a normal human ADA gene added to autologous hematopoietic stem cells (HSC) for over 30 years. Iterative improvements in vector design, HSC processing methods, and clinical HSC transplant procedures have led nearly all ADA SCID gene therapy patients to achieve consistently beneficial immune restoration with stable engraftment of ADA gene-corrected HSC over the duration of observation (as long as 20 years). One gene therapy for ADA SCID is approved by the European Medicines Agency (EMA) in the European Union (EU) and another is being advanced to licensure in the U.S. and U.K. Despite the clear-cut benefits and safety of this curative gene and cell therapy, it remains challenging to achieve sustained availability and access, especially for rare disorders like ADA SCID.


Subject(s)
Agammaglobulinemia , Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency , Humans , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/genetics , Genetic Therapy/methods
19.
Genome Res ; 34(2): 231-242, 2024 03 20.
Article in English | MEDLINE | ID: mdl-38471738

ABSTRACT

A-to-I RNA editing is a widespread epitranscriptomic phenomenon leading to the conversion of adenosines to inosines, which are primarily interpreted as guanosines by cellular machines. Consequently, A-to-I editing can alter splicing or lead to recoding of transcripts. As misregulation of editing can cause a variety of human diseases, A-to-I editing requires tight regulation of the extent of deamination, particularly in protein-coding regions. The bulk of A-to-I editing occurs cotranscriptionally. Thus, we studied A-to-I editing regulation in the context of transcription and pre-mRNA processing. We show that stimulation of transcription impacts editing levels. Activation of the transcription factor MYC leads to an up-regulation of A-to-I editing, particularly in transcripts that are suppressed upon MYC activation. Moreover, low pre-mRNA synthesis rates and low pre-mRNA expression levels support high levels of editing. We also show that editing levels greatly differ between nascent pre-mRNA and mRNA in a cellular system, as well as in mouse tissues. Editing levels can increase or decrease from pre-mRNA to mRNA and can vary across editing targets and across tissues, showing that pre-mRNA processing is an important layer of editing regulation. Several lines of evidence suggest that the differences emerge during pre-mRNA splicing. Moreover, actinomycin D treatment of primary neuronal cells and editing level analysis suggests that regulation of editing levels also depends on transcription.


Subject(s)
RNA Editing , RNA Polymerase II , RNA Precursors , Animals , Humans , Mice , Adenosine Deaminase/genetics , RNA Polymerase II/genetics , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/metabolism , Transcription, Genetic , RNA Editing/genetics , RNA Editing/physiology
SELECTION OF CITATIONS
SEARCH DETAIL