37
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      ATP hydrolysis by the viral RNA sensor RIG-I prevents unintentional recognition of self-RNA

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The cytosolic antiviral innate immune sensor RIG-I distinguishes 5′ tri- or diphosphate containing viral double-stranded (ds) RNA from self-RNA by an incompletely understood mechanism that involves ATP hydrolysis by RIG-I's RNA translocase domain. Recently discovered mutations in ATPase motifs can lead to the multi-system disorder Singleton-Merten Syndrome (SMS) and increased interferon levels, suggesting misregulated signaling by RIG-I. Here we report that SMS mutations phenocopy a mutation that allows ATP binding but prevents hydrolysis. ATPase deficient RIG-I constitutively signals through endogenous RNA and co-purifies with self-RNA even from virus infected cells. Biochemical studies and cryo-electron microscopy identify a 60S ribosomal expansion segment as a dominant self-RNA that is stably bound by ATPase deficient RIG-I. ATP hydrolysis displaces wild-type RIG-I from this self-RNA but not from 5' triphosphate dsRNA. Our results indicate that ATP-hydrolysis prevents recognition of self-RNA and suggest that SMS mutations lead to unintentional signaling through prolonged RNA binding.

          DOI: http://dx.doi.org/10.7554/eLife.10859.001

          eLife digest

          Living cells produce long, strand-like molecules of RNA that carry the instructions needed to make proteins. Viruses also make use of RNA molecules to hijack an infected cell’s protein-production machinery and create new copies of the virus. RNA molecules from viruses have a number of features that distinguish them from a cell’s own RNAs, and human cells contain receptors called RLRs that can start an immune response whenever they detect viral RNAs. All of these receptors break down molecules of ATP, a process that releases useable energy. However, so far it is not understood how this activity helps the receptors to distinguish viral RNA from the cell’s own RNA molecules (called self-RNA).

          Recently, some autoimmune diseases (including Singleton-Merten Syndrome) were linked to mutations in the parts of RLRs that allow the receptors to break down ATP. Now, Lässig et al. have studied the effects of specific mutations in an RLR called RIG-I in human cells. The experiments showed that mutations that disrupt RIG-I’s ability to bind to ATP also prevented the receptor from becoming activated. However, mutations linked to Singleton-Merten Syndrome don’t stop ATP from binding but instead slow its breakdown; this effectively locks the receptor in an ATP-bound state. Lässig et al. found that similar mutations in RIG-I caused human cells to trigger a constant immune response against the self-RNAs.

          Further experiments then suggested that the breakdown of ATP helps to remove RIG-I that has bound to double-stranded sections of self-RNAs. This activity frees the receptor, making it more able to detect double-stranded viral RNAs and preventing unintentional signaling. Lässig et al. also identified a specific double-stranded section of a human RNA that may be recognized by the mutated version of RIG-I in people with Singleton-Merten Syndrome.

          The next steps following on from this work are to extend the analysis to also include other RLRs and further explore the underlying mutations within the three-dimensional structures of the receptors and RNA molecules involved.

          DOI: http://dx.doi.org/10.7554/eLife.10859.002

          Related collections

          Most cited references43

          • Record: found
          • Abstract: found
          • Article: not found

          RIG-I-mediated antiviral responses to single-stranded RNA bearing 5'-phosphates.

          Double-stranded RNA (dsRNA) produced during viral replication is believed to be the critical trigger for activation of antiviral immunity mediated by the RNA helicase enzymes retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5). We showed that influenza A virus infection does not generate dsRNA and that RIG-I is activated by viral genomic single-stranded RNA (ssRNA) bearing 5'-phosphates. This is blocked by the influenza protein nonstructured protein 1 (NS1), which is found in a complex with RIG-I in infected cells. These results identify RIG-I as a ssRNA sensor and potential target of viral immune evasion and suggest that its ability to sense 5'-phosphorylated RNA evolved in the innate immune system as a means of discriminating between self and nonself.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Structural basis for the activation of innate immune pattern-recognition receptor RIG-I by viral RNA.

            RIG-I is a key innate immune pattern-recognition receptor that triggers interferon expression upon detection of intracellular 5'triphosphate double-stranded RNA (5'ppp-dsRNA) of viral origin. RIG-I comprises N-terminal caspase activation and recruitment domains (CARDs), a DECH helicase, and a C-terminal domain (CTD). We present crystal structures of the ligand-free, autorepressed, and RNA-bound, activated states of RIG-I. Inactive RIG-I has an open conformation with the CARDs sequestered by a helical domain inserted between the two helicase moieties. ATP and dsRNA binding induce a major rearrangement to a closed conformation in which the helicase and CTD bind the blunt end 5'ppp-dsRNA with perfect complementarity but incompatibly with continued CARD binding. We propose that after initial binding of 5'ppp-dsRNA to the flexibly linked CTD, co-operative tight binding of ATP and RNA to the helicase domain liberates the CARDs for downstream signaling. These findings significantly advance our molecular understanding of the activation of innate immune signaling helicases. Copyright © 2011 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              LGP2 is a positive regulator of RIG-I- and MDA5-mediated antiviral responses.

              RNA virus infection is recognized by retinoic acid-inducible gene (RIG)-I-like receptors (RLRs), RIG-I, and melanoma differentiation-associated gene 5 (MDA5) in the cytoplasm. RLRs are comprised of N-terminal caspase-recruitment domains (CARDs) and a DExD/H-box helicase domain. The third member of the RLR family, LGP2, lacks any CARDs and was originally identified as a negative regulator of RLR signaling. In the present study, we generated mice lacking LGP2 and found that LGP2 was required for RIG-I- and MDA5-mediated antiviral responses. In particular, LGP2 was essential for type I IFN production in response to picornaviridae infection. Overexpression of the CARDs from RIG-I and MDA5 in Lgp2(-/-) fibroblasts activated the IFN-beta promoter, suggesting that LGP2 acts upstream of RIG-I and MDA5. We further examined the role of the LGP2 helicase domain by generating mice harboring a point mutation of Lys-30 to Ala (Lgp2 (K30A/K30A)) that abrogated the LGP2 ATPase activity. Lgp2 (K30A/K30A) dendritic cells showed impaired IFN-beta productions in response to various RNA viruses to extents similar to those of Lgp2(-/-) cells. Lgp2(-/-) and Lgp2 (K30A/K30A) mice were highly susceptible to encephalomyocarditis virus infection. Nevertheless, LGP2 and its ATPase activity were dispensable for the responses to synthetic RNA ligands for MDA5 and RIG-I. Taken together, the present data suggest that LGP2 facilitates viral RNA recognition by RIG-I and MDA5 through its ATPase domain.
                Bookmark

                Author and article information

                Contributors
                Role: Reviewing editor
                Journal
                eLife
                Elife
                eLife
                eLife
                eLife
                eLife Sciences Publications, Ltd
                2050-084X
                26 November 2015
                2015
                : 4
                : e10859
                Affiliations
                [1 ]deptGene Center, Department of Biochemistry , Ludwig Maximilian University of Munich , Munich, Germany
                [2 ]deptMax von Pettenkofer-Institute, Gene Center , Ludwig Maximilian University of Munich , Munich, Germany
                [3 ]deptDepartment of Microbiology , Icahn School of Medicine at Mount Sinai , New York, United States
                [4 ]deptGlobal Health and Emerging Pathogens Institute , Icahn School of Medicine at Mount Sinai , New York, United States
                [5 ]deptInstitute for Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn , University of Bonn , Bonn, Germany
                [6 ]deptDepartment of Medicine, Division of Infectious Diseases , Icahn School of Medicine at Mount Sinai , New York, United States
                [7 ]deptInstitute of Molecular Medicine, University Hospital Bonn , University of Bonn , Bonn, Germany
                [8 ]Center for Integrated Protein Science Munich , Munich, Germany
                [9]University of California, Davis , United States
                [10]University of California, Davis , United States
                Author notes
                [‡]

                Department of Microbiology and Immunobiology, Harvard Medical School, Boston, United States.

                [†]

                These authors contributed equally to this work.

                Author information
                http://orcid.org/0000-0002-4528-8357
                Article
                10859
                10.7554/eLife.10859
                4733034
                26609812
                88caa443-49a2-408b-8b64-0c4513da8fba
                © 2015, Lässig et al

                This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

                History
                : 14 August 2015
                : 25 November 2015
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: T32 training grant 5T32AI007647-13
                Award Recipient :
                Funded by: German Excellence Initiative;
                Award ID: CIPSM
                Award Recipient :
                Funded by: Graduate School of Quantitative Biosciences Munich;
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100000781, European Research Council;
                Award ID: Advanced Grant CRYOTRANSLATION
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001659, Deutsche Forschungsgemeinschaft;
                Award ID: FOR 1805
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001659, Deutsche Forschungsgemeinschaft;
                Award ID: SFB646
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100001659, Deutsche Forschungsgemeinschaft;
                Award ID: GRK1721
                Award Recipient :
                Funded by: Bavarian network for Molecular Biosystems;
                Award Recipient :
                The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication.
                Categories
                Research Article
                Cell Biology
                Immunology
                Custom metadata
                2.5
                Mutations within the ATPase domain of RIG-I in patients with Singleton-Merten Syndrome prevent ATP-hydrolysis dependent dissociation of RIG-I from double-stranded RNA and lead to unintentional constitutive signaling through increased binding of endogenous RNA.

                Life sciences
                rig-i,rlr,innate immune system,atpase domain,autoimmune response / disease,singleton-merten syndrome,human

                Comments

                Comment on this article