Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 635
Filter
Add more filters

Publication year range
1.
Cell ; 185(19): 3588-3602.e21, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36113429

ABSTRACT

The current dogma of RNA-mediated innate immunity is that sensing of immunostimulatory RNA ligands is sufficient for the activation of intracellular sensors and induction of interferon (IFN) responses. Here, we report that actin cytoskeleton disturbance primes RIG-I-like receptor (RLR) activation. Actin cytoskeleton rearrangement induced by virus infection or commonly used reagents to intracellularly deliver RNA triggers the relocalization of PPP1R12C, a regulatory subunit of the protein phosphatase-1 (PP1), from filamentous actin to cytoplasmic RLRs. This allows dephosphorylation-mediated RLR priming and, together with the RNA agonist, induces effective RLR downstream signaling. Genetic ablation of PPP1R12C impairs antiviral responses and enhances susceptibility to infection with several RNA viruses including SARS-CoV-2, influenza virus, picornavirus, and vesicular stomatitis virus. Our work identifies actin cytoskeleton disturbance as a priming signal for RLR-mediated innate immunity, which may open avenues for antiviral or adjuvant design.


Subject(s)
Actins , COVID-19 , Actin Cytoskeleton , Antiviral Agents , Humans , Interferons , Ligands , Protein Phosphatase 1 , RNA , RNA Helicases , Receptors, Retinoic Acid/metabolism , SARS-CoV-2
2.
Cell ; 184(19): 4981-4995.e14, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34464586

ABSTRACT

Poor tumor infiltration, development of exhaustion, and antigen insufficiency are common mechanisms that limit chimeric antigen receptor (CAR)-T cell efficacy. Delivery of pattern recognition receptor agonists is one strategy to improve immune function; however, targeting these agonists to immune cells is challenging, and off-target signaling in cancer cells can be detrimental. Here, we engineer CAR-T cells to deliver RN7SL1, an endogenous RNA that activates RIG-I/MDA5 signaling. RN7SL1 promotes expansion and effector-memory differentiation of CAR-T cells. Moreover, RN7SL1 is deployed in extracellular vesicles and selectively transferred to immune cells. Unlike other RNA agonists, transferred RN7SL1 restricts myeloid-derived suppressor cell (MDSC) development, decreases TGFB in myeloid cells, and fosters dendritic cell (DC) subsets with costimulatory features. Consequently, endogenous effector-memory and tumor-specific T cells also expand, allowing rejection of solid tumors with CAR antigen loss. Supported by improved endogenous immunity, CAR-T cells can now co-deploy peptide antigens with RN7SL1 to enhance efficacy, even when heterogenous CAR antigen tumors lack adequate neoantigens.


Subject(s)
Immunologic Factors/pharmacology , RNA/pharmacology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , DEAD Box Protein 58/metabolism , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Extracellular Vesicles/metabolism , Humans , Immunity/drug effects , Immunocompetence , Immunologic Memory , Immunotherapy , Interferons/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Peptides/metabolism , Receptors, Pattern Recognition/metabolism , T-Lymphocytes/drug effects
3.
Immunity ; 57(4): 731-751, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38599168

ABSTRACT

RIG-I-like receptors (RLRs) are crucial for pathogen detection and triggering immune responses and have immense physiological importance. In this review, we first summarize the interferon system and innate immunity, which constitute primary and secondary responses. Next, the molecular structure of RLRs and the mechanism of sensing non-self RNA are described. Usually, self RNA is refractory to the RLR; however, there are underlying host mechanisms that prevent immune reactions. Studies have revealed that the regulatory mechanisms of RLRs involve covalent molecular modifications, association with regulatory factors, and subcellular localization. Viruses have evolved to acquire antagonistic RLR functions to escape the host immune reactions. Finally, the pathologies caused by the malfunction of RLR signaling are described.


Subject(s)
DEAD-box RNA Helicases , Signal Transduction , DEAD-box RNA Helicases/genetics , Interferon-Induced Helicase, IFIH1/metabolism , DEAD Box Protein 58 , Immunity, Innate , Receptors, Immunologic , RNA
4.
Cell ; 172(4): 797-810.e13, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29395326

ABSTRACT

Aberrant activation of innate immune receptors can cause a spectrum of immune disorders, such as Aicardi-Goutières syndrome (AGS). One such receptor is MDA5, a viral dsRNA sensor that induces antiviral immune response. Using a newly developed RNase-protection/RNA-seq approach, we demonstrate here that constitutive activation of MDA5 in AGS results from the loss of tolerance to cellular dsRNAs formed by Alu retroelements. While wild-type MDA5 cannot efficiently recognize Alu-dsRNAs because of its limited filament formation on imperfect duplexes, AGS variants of MDA5 display reduced sensitivity to duplex structural irregularities, assembling signaling-competent filaments on Alu-dsRNAs. Moreover, we identified an unexpected role of an RNA-rich cellular environment in suppressing aberrant MDA5 oligomerization, highlighting context dependence of self versus non-self discrimination. Overall, our work demonstrates that the increased efficiency of MDA5 in recognizing dsRNA comes at a cost of self-recognition and implicates a unique role of Alu-dsRNAs as virus-like elements that shape the primate immune system.


Subject(s)
Alu Elements/immunology , Autoimmune Diseases of the Nervous System/immunology , Interferon-Induced Helicase, IFIH1/immunology , Nervous System Malformations/immunology , Protein Multimerization/immunology , RNA, Double-Stranded/immunology , Self Tolerance , A549 Cells , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/pathology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interferon-Induced Helicase, IFIH1/genetics , Muramidase , Nervous System Malformations/genetics , Nervous System Malformations/pathology , Peptide Fragments , Protein Multimerization/genetics , RNA, Double-Stranded/genetics , THP-1 Cells
5.
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
6.
Mol Cell ; 84(1): 107-119, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38118451

ABSTRACT

The ability to sense and respond to infection is essential for life. Viral infection produces double-stranded RNAs (dsRNAs) that are sensed by proteins that recognize the structure of dsRNA. This structure-based recognition of viral dsRNA allows dsRNA sensors to recognize infection by many viruses, but it comes at a cost-the dsRNA sensors cannot always distinguish between "self" and "nonself" dsRNAs. "Self" RNAs often contain dsRNA regions, and not surprisingly, mechanisms have evolved to prevent aberrant activation of dsRNA sensors by "self" RNA. Here, we review current knowledge about the life of endogenous dsRNAs in mammals-the biosynthesis and processing of dsRNAs, the proteins they encounter, and their ultimate degradation. We highlight mechanisms that evolved to prevent aberrant dsRNA sensor activation and the importance of competition in the regulation of dsRNA sensors and other dsRNA-binding proteins.


Subject(s)
RNA, Double-Stranded , Virus Diseases , Animals , RNA, Double-Stranded/genetics , DEAD-box RNA Helicases/metabolism , Immunity, Innate , Mammals/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(10): 2218-2230.e5, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34644557

ABSTRACT

The RNA sensor MDA5 recruits the signaling adaptor MAVS to initiate type I interferon signaling and downstream antiviral responses, a process that requires K63-linked polyubiquitin chains. Here, we examined the mechanisms whereby K63-polyUb chain regulate MDA5 activation. Only long unanchored K63-polyUbn (n ≥ 8) could mediate tetramerization of the caspase activation and recruitment domains of MDA5 (MDA5CARDs). Cryoelectron microscopy structures of a polyUb13-bound MDA5CARDs tetramer and a polyUb11-bound MDA5CARDs-MAVSCARD assembly revealed a tower-like formation, wherein eight Ubs tethered along the outer rim of the helical shell, bridging MDA5CARDs and MAVSCARD tetramers into proximity. ATP binding and hydrolysis promoted the stabilization of RNA-bound MDA5 prior to MAVS activation via allosteric effects on CARDs-polyUb complex. Abundant ATP prevented basal activation of apo MDA5. Our findings reveal the ordered assembly of a MDA5 signaling complex competent to recruit and activate MAVS and highlight differences with RIG-I in terms of CARD orientation and Ub sensing that suggest different abilities to induce antiviral responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/chemistry , Cryoelectron Microscopy , HEK293 Cells , Humans , Immunity, Innate/physiology , Interferon-Induced Helicase, IFIH1/chemistry , Interferon-Induced Helicase, IFIH1/ultrastructure , Polyubiquitin/chemistry , Polyubiquitin/metabolism , Protein Binding
9.
Immunity ; 54(9): 1948-1960.e5, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34343497

ABSTRACT

The RNA deaminase ADAR1 is an essential negative regulator of the RNA sensor MDA5, and loss of ADAR1 function triggers inappropriate activation of MDA5 by self-RNAs. Mutations in ADAR, the gene that encodes ADAR1, cause human immune diseases, including Aicardi-Goutières syndrome (AGS). However, the mechanisms of MDA5-dependent disease pathogenesis in vivo remain unknown. Here we generated mice with a single amino acid change in ADAR1 that models the most common human ADAR AGS mutation. These Adar mutant mice developed lethal disease that required MDA5, the RIG-I-like receptor LGP2, type I interferons, and the eIF2α kinase PKR. A small-molecule inhibitor of the integrated stress response (ISR) that acts downstream of eIF2α phosphorylation prevented immunopathology and rescued the mice from mortality. These findings place PKR and the ISR as central components of immunopathology in vivo and identify therapeutic targets for treatment of human diseases associated with the ADAR1-MDA5 axis.


Subject(s)
Adenosine Deaminase/metabolism , Autoimmune Diseases of the Nervous System/pathology , Nervous System Malformations/pathology , Stress, Physiological/physiology , eIF-2 Kinase/metabolism , A549 Cells , Animals , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/metabolism , Disease Models, Animal , HEK293 Cells , Humans , Mice , Mice, Mutant Strains , Mutation , Nervous System Malformations/genetics , Nervous System Malformations/metabolism
10.
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
11.
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
12.
Mol Cell ; 81(3): 599-613.e8, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33373584

ABSTRACT

RNA helicases and E3 ubiquitin ligases mediate many critical functions in cells, but their actions have largely been studied in distinct biological contexts. Here, we uncover evolutionarily conserved rules of engagement between RNA helicases and tripartite motif (TRIM) E3 ligases that lead to their functional coordination in vertebrate innate immunity. Using cryoelectron microscopy and biochemistry, we show that RIG-I-like receptors (RLRs), viral RNA receptors with helicase domains, interact with their cognate TRIM/TRIM-like E3 ligases through similar epitopes in the helicase domains. Their interactions are avidity driven, restricting the actions of TRIM/TRIM-like proteins and consequent immune activation to RLR multimers. Mass spectrometry and phylogeny-guided biochemical analyses further reveal that similar rules of engagement may apply to diverse RNA helicases and TRIM/TRIM-like proteins. Our analyses suggest not only conserved substrates for TRIM proteins but also, unexpectedly, deep evolutionary connections between TRIM proteins and RNA helicases, linking ubiquitin and RNA biology throughout animal evolution.


Subject(s)
DEAD Box Protein 58/metabolism , Immunity, Innate , Interferon-Induced Helicase, IFIH1/metabolism , Receptors, Immunologic/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Cryoelectron Microscopy , DEAD Box Protein 58/genetics , DEAD Box Protein 58/ultrastructure , Epitopes , Evolution, Molecular , HEK293 Cells , Humans , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/ultrastructure , Models, Molecular , Phylogeny , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Receptors, Immunologic/genetics , Receptors, Immunologic/ultrastructure , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/ultrastructure , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/ultrastructure
13.
Trends Genet ; 40(3): 250-259, 2024 03.
Article in English | MEDLINE | ID: mdl-38160061

ABSTRACT

Recent studies have underscored the pivotal role of adenosine-to-inosine RNA editing, catalyzed by ADAR1, in suppressing innate immune interferon responses triggered by cellular double-stranded RNA (dsRNA). However, the specific ADAR1 editing targets crucial for this regulatory function remain elusive. We review analyses of transcriptome-wide ADAR1 editing patterns and their evolutionary dynamics, which offer valuable insights into this unresolved query. The growing appreciation of the significance of immunogenic dsRNAs and their editing in inflammatory and autoimmune diseases and cancer calls for a more comprehensive understanding of dsRNA immunogenicity, which may promote our understanding of these diseases and open doors to therapeutic avenues.


Subject(s)
Autoimmune Diseases , RNA, Double-Stranded , Humans , RNA, Double-Stranded/genetics , Immunity, Innate/genetics , Transcriptome/genetics
14.
Proc Natl Acad Sci U S A ; 121(29): e2404349121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38985764

ABSTRACT

Intron-containing RNA expressed from the HIV-1 provirus activates type 1 interferon in primary human blood cells, including CD4+ T cells, macrophages, and dendritic cells. To identify the innate immune receptor required for detection of intron-containing RNA expressed from the HIV-1 provirus, a loss-of-function screen was performed with short hairpin RNA-expressing lentivectors targeting twenty-one candidate genes in human monocyte-derived dendritic cells. Among the candidate genes tested, only knockdown of XPO1 (CRM1), IFIH1 (MDA5), or MAVS prevented activation of the interferon-stimulated gene ISG15. The importance of IFIH1 protein was demonstrated by rescue of the knockdown with nontargetable IFIH1 coding sequence. Inhibition of HIV-1-induced ISG15 by the IFIH1-specific Nipah virus V protein, and by IFIH1-transdominant 2-CARD domain-deletion or phosphomimetic point mutations, indicates that IFIH1 (MDA5) filament formation, dephosphorylation, and association with MAVS are all required for innate immune activation in response to HIV-1 transduction. Since both IFIH1 (MDA5) and DDX58 (RIG-I) signal via MAVS, the specificity of HIV-1 RNA detection by IFIH1 was demonstrated by the fact that DDX58 knockdown had no effect on activation. RNA-Seq showed that IFIH1 knockdown in dendritic cells globally disrupted the induction of IFN-stimulated genes by HIV-1. Finally, specific enrichment of unspliced HIV-1 RNA by IFIH1 (MDA5), over two orders of magnitude, was revealed by formaldehyde cross-linking immunoprecipitation (f-CLIP). These results demonstrate that IFIH1 is the innate immune receptor for intron-containing RNA from the HIV-1 provirus and that IFIH1 potentially contributes to chronic inflammation in people living with HIV-1, even in the presence of effective antiretroviral therapy.


Subject(s)
Dendritic Cells , HIV-1 , Immunity, Innate , Interferon-Induced Helicase, IFIH1 , Introns , Proviruses , RNA, Viral , Humans , HIV-1/genetics , HIV-1/immunology , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Proviruses/genetics , Dendritic Cells/immunology , Dendritic Cells/virology , Dendritic Cells/metabolism , Introns/genetics , RNA, Viral/genetics , RNA, Viral/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/immunology , HIV Infections/immunology , HIV Infections/virology , HIV Infections/genetics , Karyopherins/genetics , Karyopherins/metabolism
15.
RNA ; 30(5): 500-511, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38531645

ABSTRACT

Innate immunity must be tightly regulated to enable sensitive pathogen detection while averting autoimmunity triggered by pathogen-like host molecules. A hallmark of viral infection, double-stranded RNAs (dsRNAs) are also abundantly encoded in mammalian genomes, necessitating surveillance mechanisms to distinguish "self" from "nonself." ADAR1, an RNA editing enzyme, has emerged as an essential safeguard against dsRNA-induced autoimmunity. By converting adenosines to inosines (A-to-I) in long dsRNAs, ADAR1 covalently marks endogenous dsRNAs, thereby blocking the activation of the cytoplasmic dsRNA sensor MDA5. Moreover, beyond its editing function, ADAR1 binding to dsRNA impedes the activation of innate immune sensors PKR and ZBP1. Recent landmark studies underscore the utility of silencing ADAR1 for cancer immunotherapy, by exploiting the ADAR1-dependence developed by certain tumors to unleash an antitumor immune response. In this perspective, we summarize the genetic and mechanistic evidence for ADAR1's multipronged role in suppressing dsRNA-mediated autoimmunity and explore the evolving roles of ADAR1 as an immuno-oncology target.


Subject(s)
Adenosine Deaminase , RNA Editing , Animals , Adenosine Deaminase/metabolism , Immunity, Innate/genetics , Interferon-Induced Helicase, IFIH1/genetics , Mammals/genetics , RNA, Double-Stranded/genetics , Humans
16.
EMBO Rep ; 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285245

ABSTRACT

As a RIG-I-like receptor, MDA5 plays a critical role in antiviral innate immunity by acting as a cytoplasmic double-stranded RNA sensor capable of initiating type I interferon pathways. Here, we show that RNF144B specifically interacts with MDA5 and promotes K27/K33-linked polyubiquitination of MDA5 at lysine 23 and lysine 43, which promotes autophagic degradation of MDA5 by p62. Rnf144b deficiency greatly promotes IFN production and inhibits EMCV replication in vivo. Importantly, Rnf144b-/- mice has a significantly higher overall survival rate than wild-type mice upon EMCV infection. Collectively, our results identify RNF144B as a negative regulator of innate antiviral response by targeting CARDs of MDA5 and mediating autophagic degradation of MDA5.

17.
Proc Natl Acad Sci U S A ; 120(36): e2214956120, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37639603

ABSTRACT

The cytosolic RNA and DNA sensors initiate type I interferon signaling when binding to RNA or DNA. To effectively protect the host against virus infection and concomitantly avoid excessive interferonopathy at resting states, these sensors must be tightly regulated. However, the key molecular mechanisms regulating these sensors' activation remain elusive. Here, we identify PRMT3, a type I protein arginine methyltransferase, as a negative regulator of cytosolic RNA and DNA sensors. PRMT3 interacts with RIG-I, MDA5, and cGAS and catalyzes asymmetric dimethylation of R730 on RIG-I, R822 on MDA5, and R111 on cGAS. These modifications reduce RNA-binding ability of RIG-I and MDA5 as well as DNA-binding ability and oligomerization of cGAS, leading to the inhibition of downstream type I interferon production. Furthermore, mice with loss of one copy of Prmt3 or in vivo treatment of the PRMT3 inhibitor, SGC707, are more resistant to RNA and DNA virus infection. Our findings reveal an essential role of PRMT3 in the regulation of antiviral innate immunity and give insights into the molecular regulation of cytosolic RNA and DNA sensors' activation.


Subject(s)
Arginine , Interferon Type I , Animals , Mice , RNA/genetics , Antiviral Agents/pharmacology , Immunity, Innate , DNA/genetics , Nucleotidyltransferases/genetics , Protein-Arginine N-Methyltransferases/genetics
18.
J Virol ; 98(3): e0000324, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38353538

ABSTRACT

The microtubule (MT) is a highly dynamic polymer that functions in various cellular processes through MT hyperacetylation. Thus, many viruses have evolved mechanisms to hijack the MT network of the cytoskeleton to allow intracellular replication of viral genomic material. Coronavirus non-structural protein 8 (nsp8), a component of the viral replication transcriptional complex, is essential for viral survival. Here, we found that nsp8 of porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus with a zoonotic potential, inhibits interferon (IFN)-ß production by targeting melanoma differentiation gene 5 (MDA5), the main pattern recognition receptor for coronaviruses in the cytoplasm. Mechanistically, PDCoV nsp8 interacted with MDA5 and induced autophagy to degrade MDA5 in wild-type cells, but not in autophagy-related (ATG)5 or ATG7 knockout cells. Further screening for autophagic degradation receptors revealed that nsp8 interacts with sequestosome 1/p62 and promotes p62-mediated selective autophagy to degrade MDA5. Importantly, PDCoV nsp8 induced hyperacetylation of MTs, which in turn triggered selective autophagic degradation of MDA5 and subsequent inhibition of IFN-ß production. Overall, our study uncovers a novel mechanism employed by PDCoV nsp8 to evade host innate immune defenses. These findings offer new insights into the interplay among viruses, IFNs, and MTs, providing a promising target to develop anti-viral drugs against PDCoV.IMPORTANCECoronavirus nsp8, a component of the viral replication transcriptional complex, is well conserved and plays a crucial role in viral replication. Exploration of the role mechanism of nsp8 is conducive to the understanding of viral pathogenesis and development of anti-viral strategies against coronavirus. Here, we found that nsp8 of PDCoV, an emerging enteropathogenic coronavirus with a zoonotic potential, is an interferon antagonist. Further studies showed that PDCoV nsp8 interacted with MDA5 and sequestosome 1/p62, promoting p62-mediated selective autophagy to degrade MDA5. We further found that PDCoV nsp8 could induce hyperacetylation of MT, therefore triggering selective autophagic degradation of MDA5 and inhibiting IFN-ß production. These findings reveal a novel immune evasion strategy used by PDCoV nsp8 and provide insights into potential therapeutic interventions.


Subject(s)
Coronavirus Infections , Deltacoronavirus , Swine Diseases , Animals , Autophagy , Coronavirus Infections/metabolism , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Deltacoronavirus/metabolism , Interferons/metabolism , Microtubules/metabolism , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Swine , Swine Diseases/virology
19.
EMBO Rep ; 24(5): e55835, 2023 05 04.
Article in English | MEDLINE | ID: mdl-36975179

ABSTRACT

ADAR1 -mediated A-to-I RNA editing is a self-/non-self-discrimination mechanism for cellular double-stranded RNAs. ADAR mutations are one cause of Aicardi-Goutières Syndrome, an inherited paediatric encephalopathy, classed as a "Type I interferonopathy." The most common ADAR1 mutation is a proline 193 alanine (p.P193A) mutation, mapping to the ADAR1p150 isoform-specific Zα domain. Here, we report the development of an independent murine P195A knock-in mouse, homologous to human P193A. The Adar1P195A/P195A mice are largely normal and the mutation is well tolerated. When the P195A mutation is compounded with an Adar1 null allele (Adar1P195A/- ), approximately half the animals are runted with a shortened lifespan while the remaining Adar1P195A/- animals are normal, contrasting with previous reports. The phenotype of the Adar1P195A/- animals is both associated with the parental genotype and partly non-genetic/environmental. Complementation with an editing-deficient ADAR1 (Adar1P195A/E861A ), or the loss of MDA5, rescues phenotypes in the Adar1P195A/- mice.


Subject(s)
RNA Editing , RNA, Double-Stranded , Humans , Mice , Animals , Child , Phenotype , Mutation , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism
20.
Biochem Biophys Res Commun ; 733: 150725, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39317111

ABSTRACT

Junctophilin-2 (JPH2) is traditionally recognized as a cardiomyocyte-enriched structural protein that anchors the junction between the plasma membrane and the endo/sarcoplasmic reticulum, facilitating excitation-induced cardiac contraction. In this study, we uncover a novel function of JPH2 as a double-stranded RNA (dsRNA)-binding protein, which forms complexes with dsRNA both in vitro and in cells. Stimulation by cytosolic dsRNA enhances the interaction of JPH2 with the dsRNA sensor MDA5. Notably, JPH2 inhibits MDA5's binding to its dsRNA ligand, likely by sequestering the dsRNA. Silencing JPH2 in cardiomyocytes increased the interaction between MDA5 and its dsRNA ligands, activated the MAVS/TBK1 signaling, and triggered spontaneous interferon-beta (IFNb1) production in the absence of foreign pathogen. Mouse hearts deficient in JPH2 exhibited upregulation of innate immune signaling cascade. Collectively, these findings identify JPH2 as a regulator of dsRNA sensing and highlight its role in suppressing the automatic activation of innate immune responses in cardiomyocytes, suggesting the cytosolic surface of the endo/sarcoplasmic reticulum as a hub for dsRNA sequestration.

SELECTION OF CITATIONS
SEARCH DETAIL