Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 439
Filter
1.
Mol Immunol ; 170: 156-169, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692097

ABSTRACT

Type-I and -III interferons play a central role in immune rejection of pathogens and tumors, thus promoting immunogenicity and suppressing tumor recurrence. Double strand RNA is an important ligand that stimulates tumor immunity via interferon responses. Differentiation of embryonic stem cells to pluripotent epithelial cells activates the interferon response during development, raising the question of whether epithelial vs. mesenchymal gene signatures in cancer potentially regulate the interferon pathway as well. Here, using genomics and signaling approaches, we show that Grainyhead-like-2 (GRHL2), a master programmer of epithelial cell identity, promotes type-I and -III interferon responses to double-strand RNA. GRHL2 enhanced the activation of IRF3 and relA/NF-kB and the expression of IRF1; a functional GRHL2 binding site in the IFNL1 promoter was also identified. Moreover, time to recurrence in breast cancer correlated positively with GRHL2 protein expression, indicating that GRHL2 is a tumor recurrence suppressor, consistent with its enhancement of interferon responses. These observations demonstrate that epithelial cell identity supports interferon responses in the context of cancer.


Subject(s)
Breast Neoplasms , DNA-Binding Proteins , Transcription Factors , Humans , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Female , Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Neoplasm Recurrence, Local/immunology , Interferons/metabolism , Interferons/immunology , Interferons/genetics , Cell Line, Tumor , Epithelial Cells/immunology , Epithelial Cells/metabolism , Animals , RNA, Double-Stranded/immunology , Transcription Factor RelA/metabolism , Mice , Gene Expression Regulation, Neoplastic , Signal Transduction/immunology , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/immunology
2.
Clin Rheumatol ; 43(3): 1111-1126, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38182800

ABSTRACT

BACKGROUND: Melanoma differentiation-associated gene 5 (MDA5), as a cytoplasmic sensor for viral double-stranded RNAs, has received increasing attention in recent years. Although considerable headway has been made on the functional role of MDA5 in antiviral immunity and autoimmune disease, the available literature is insufficient to assess the vast field. METHODS: This study performed a bibliometric analysis to investigate current hotspots in the global scientific output of MDA5 over the past two decades. Related publications and recorded information from 2002 to 2022 in the Web of Science Core Collection (WoSCC) database were retrieved. VOSviewer and CiteSpace were used for quantitative evaluation and visualization. RESULTS: A total of 2267 original articles and reviews were obtained, and the annual number of publications related to MDA5 was increasing rapidly. China has published the most papers, while the USA was the most influential country with the most citations and the highest H-index. The Chinese Academy of Sciences, the United States Department of Health and Human Services, and the Journal of Virology were the most prolific research affiliation, funding source, and journal, respectively. Fujita T (Kyoto University) was the most productive author with the highest H-index and had close cooperation with Kato H and Yoneyama M. The keywords "RIG-I," "MDA5," "innate immunity," "double-stranded-RNA," and "recognition" had the highest frequency, while "dermatomyositis" as well as "autoantibody" seemed to be the emerging hotspots. CONCLUSION: This study comprehensively demonstrated the research frontiers of MDA5 and will provide a useful resource for scholars to conduct future decisions. KEY POINTS: We conducted the first in-depth survey of the research frontiers on melanoma differentiation-associated gene 5 (MDA5) over the past two decades via bibliometric analysis. We found that many early breakthroughs have been made in the mechanism of MDA5-mediated antiviral immune responses, and the role of MDA5 in autoimmune and autoinflammatory diseases has raised the recent concern. We identified that the virus infection-associated pathogenesis and effective therapeutic strategy of anti-MDA5 antibody-positive dermatomyositis will remain the hotspots in the future.


Subject(s)
Autoimmune Diseases , Interferon-Induced Helicase, IFIH1 , RNA, Viral , Humans , Autoantibodies/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/virology , Bibliometrics , China , Double Stranded RNA Viruses/immunology , Interferon-Induced Helicase, IFIH1/immunology , RNA, Double-Stranded/immunology , RNA, Viral/immunology , United States
3.
Nature ; 617(7962): 818-826, 2023 05.
Article in English | MEDLINE | ID: mdl-37198486

ABSTRACT

Cancer cells rewire metabolism to favour the generation of specialized metabolites that support tumour growth and reshape the tumour microenvironment1,2. Lysine functions as a biosynthetic molecule, energy source and antioxidant3-5, but little is known about its pathological role in cancer. Here we show that glioblastoma stem cells (GSCs) reprogram lysine catabolism through the upregulation of lysine transporter SLC7A2 and crotonyl-coenzyme A (crotonyl-CoA)-producing enzyme glutaryl-CoA dehydrogenase (GCDH) with downregulation of the crotonyl-CoA hydratase enoyl-CoA hydratase short chain 1 (ECHS1), leading to accumulation of intracellular crotonyl-CoA and histone H4 lysine crotonylation. A reduction in histone lysine crotonylation by either genetic manipulation or lysine restriction impaired tumour growth. In the nucleus, GCDH interacts with the crotonyltransferase CBP to promote histone lysine crotonylation. Loss of histone lysine crotonylation promotes immunogenic cytosolic double-stranded RNA (dsRNA) and dsDNA generation through enhanced H3K27ac, which stimulates the RNA sensor MDA5 and DNA sensor cyclic GMP-AMP synthase (cGAS) to boost type I interferon signalling, leading to compromised GSC tumorigenic potential and elevated CD8+ T cell infiltration. A lysine-restricted diet synergized with MYC inhibition or anti-PD-1 therapy to slow tumour growth. Collectively, GSCs co-opt lysine uptake and degradation to shunt the production of crotonyl-CoA, remodelling the chromatin landscape to evade interferon-induced intrinsic effects on GSC maintenance and extrinsic effects on immune response.


Subject(s)
Histones , Lysine , Neoplasms , Protein Processing, Post-Translational , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Histones/chemistry , Histones/metabolism , Lysine/deficiency , Lysine/metabolism , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , RNA, Double-Stranded/immunology , Humans , Animals , Mice , Interferon Type I/immunology
4.
J Immunol ; 210(3): 335-347, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36525065

ABSTRACT

Melanoma differentiation-associated gene 5 (MDA5), a member of the retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), has pivotal roles in innate immune responses against many positive-stranded RNA viruses, including picornavirus and coronavirus. Upon engagement with dsRNA derived from viral infection, MDA5 initiates coordinated signal transduction leading to type I IFN induction to restrict viral replication. In this study, we describe a targeted cleavage events of MDA5 by the 3C protease from Theilovirus. Upon ectopic expression of theilovirus 3C protease from Saffold virus or Theiler's murine encephalomyelitis virus but not encephalomyocarditis virus, fragments of cleaved MDA5 were observed in a dose-dependent manner. When enzymatically inactive Theilovirus 3C protease was expressed, MDA5 cleavage was completely abrogated. Mass spectrometric analysis identified two cleavage sites at the C terminus of MDA5, cleaving off one of the RNA-binding domains. The same cleavage pattern was observed during Theilovirus infection. The cleavage of MDA5 by Theilovirus protease impaired ATP hydrolysis, RNA binding, and filament assembly on RNA, resulting in dysfunction of MDA5 as an innate immune RNA sensor for IFN induction. Furthermore, the cleavage-resistant MDA5 mutant against the 3C protease showed an enhanced IFN response during Saffold virus infection, indicating that Theilovirus has a strategy to circumvent the antiviral immune response by cleaving MDA5 using 3C protease. In summary, these data suggest MDA5 cleavage by 3C protease as a novel immune evasive strategy of Theilovirus.


Subject(s)
Interferon-Induced Helicase, IFIH1 , RNA, Double-Stranded , Theilovirus , Animals , Mice , Cysteine Endopeptidases/genetics , Host-Pathogen Interactions , Immunity, Innate , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Peptide Hydrolases/metabolism , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , 3C Viral Proteases
5.
Proc Natl Acad Sci U S A ; 119(33): e2204235119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35939694

ABSTRACT

Mammalian cells respond to dsRNA in multiple manners. One key response to dsRNA is the activation of PKR, an eIF2α kinase, which triggers translational arrest and the formation of stress granules. However, the process of PKR activation in cells is not fully understood. In response to increased endogenous or exogenous dsRNA, we observed that PKR forms novel cytosolic condensates, referred to as dsRNA-induced foci (dRIFs). dRIFs contain dsRNA, form in proportion to dsRNA, and are enhanced by longer dsRNAs. dRIFs enrich several other dsRNA-binding proteins, including ADAR1, Stau1, NLRP1, and PACT. Strikingly, dRIFs correlate with and form before translation repression by PKR and localize to regions of cells where PKR activation is initiated. We hypothesize that dRIF formation is a mechanism that cells use to enhance the sensitivity of PKR activation in response to low levels of dsRNA or to overcome viral inhibitors of PKR activation.


Subject(s)
RNA, Double-Stranded , RNA, Viral , Virus Diseases , eIF-2 Kinase , Enzyme Activation , Humans , Immunity, Innate , Phosphorylation , Protein Biosynthesis , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/immunology , RNA, Viral/chemistry , RNA, Viral/immunology , RNA-Binding Proteins/chemistry , Stress Granules , Virus Diseases/enzymology , Virus Diseases/immunology , eIF-2 Kinase/chemistry
6.
Nature ; 606(7914): 594-602, 2022 06.
Article in English | MEDLINE | ID: mdl-35614224

ABSTRACT

Only a small proportion of patients with cancer show lasting responses to immune checkpoint blockade (ICB)-based monotherapies. The RNA-editing enzyme ADAR1 is an emerging determinant of resistance to ICB therapy and prevents ICB responsiveness by repressing immunogenic double-stranded RNAs (dsRNAs), such as those arising from the dysregulated expression of endogenous retroviral elements (EREs)1-4. These dsRNAs trigger an interferon-dependent antitumour response by activating A-form dsRNA (A-RNA)-sensing proteins such as MDA-5 and PKR5. Here we show that ADAR1 also prevents the accrual of endogenous Z-form dsRNA elements (Z-RNAs), which were enriched in the 3' untranslated regions of interferon-stimulated mRNAs. Depletion or mutation of ADAR1 resulted in Z-RNA accumulation and activation of the Z-RNA sensor ZBP1, which culminated in RIPK3-mediated necroptosis. As no clinically viable ADAR1 inhibitors currently exist, we searched for a compound that can override the requirement for ADAR1 inhibition and directly activate ZBP1. We identified a small molecule, the curaxin CBL0137, which potently activates ZBP1 by triggering Z-DNA formation in cells. CBL0137 induced ZBP1-dependent necroptosis in cancer-associated fibroblasts and reversed ICB unresponsiveness in mouse models of melanoma. Collectively, these results demonstrate that ADAR1 represses endogenous Z-RNAs and identifies ZBP1-mediated necroptosis as a new determinant of tumour immunogenicity masked by ADAR1. Therapeutic activation of ZBP1-induced necroptosis provides a readily translatable avenue for rekindling the immune responsiveness of ICB-resistant human cancers.


Subject(s)
Adenosine Deaminase , Necroptosis , Neoplasms , RNA-Binding Proteins , 3' Untranslated Regions , Adenosine Deaminase/metabolism , Animals , Cancer-Associated Fibroblasts , Carbazoles/pharmacology , Humans , Immunotherapy/trends , Interferons/metabolism , Melanoma , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , RNA, Double-Stranded/immunology , RNA-Binding Proteins/metabolism
7.
Front Immunol ; 13: 818023, 2022.
Article in English | MEDLINE | ID: mdl-35126398

ABSTRACT

Alu retrotransposons belong to the class of short interspersed nuclear elements (SINEs). Alu RNA is abundant in cells and its repetitive structure forms double-stranded RNAs (dsRNA) that activate dsRNA sensors and trigger innate immune responses with significant pathological consequences. Mechanisms to prevent innate immune activation include deamination of adenosines to inosines in dsRNAs, referred to as A-to-I editing, degradation of Alu RNAs by endoribonucleases, and sequestration of Alu RNAs by RNA binding proteins. We have previously demonstrated that widespread loss of Alu RNA A-to-I editing is associated with diverse human diseases including viral (COVID-19, influenza) and autoimmune diseases (multiple sclerosis). Here we demonstrate loss of A-to-I editing in leukocytes is also associated with inflammatory bowel diseases. Our structure-function analysis demonstrates that ability to activate innate immune responses resides in the left arm of Alu RNA, requires a 5'-PPP, RIG-I is the major Alu dsRNA sensor, and A-to-I editing disrupts both structure and function. Further, edited Alu RNAs inhibit activity of unedited Alu RNAs. Altering Alu RNA nucleotide sequence increases biological activity. Two classes of Alu RNAs exist, one class stimulates both IRF and NF-kB transcriptional activity and a second class only stimulates IRF transcriptional activity. Thus, Alu RNAs play important roles in human disease but may also have therapeutic potential.


Subject(s)
Alu Elements/genetics , Alu Elements/immunology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Adenosine , COVID-19 , Humans , Inosine , RNA, Double-Stranded/genetics , RNA, Double-Stranded/immunology , SARS-CoV-2
8.
Viruses ; 14(1)2022 01 05.
Article in English | MEDLINE | ID: mdl-35062293

ABSTRACT

The physiologic function of tripartite motif protein 56 (TRIM56), a ubiquitously expressed E3 ligase classified within the large TRIM protein family, remains elusive. Gene knockdown studies have suggested TRIM56 as a positive regulator of the type I interferon (IFN-I) antiviral response elicited via the Toll-like receptor 3 (TLR3) and cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathways, which detect and respond to danger signals-extracellular double-stranded (ds) RNA and cytosolic dsDNA, respectively. However, to what extent these pathways depend on TRIM56 in human cells is unclear. In addition, it is debatable whether TRIM56 plays a part in controlling the expression of IFN-stimulated genes (ISGs) resulting from IFN-I based antiviral treatment. In this study, we created HeLa-derived TRIM56 null cell lines by gene editing and used these cell models to comprehensively examine the impact of endogenous TRIM56 on innate antiviral responses. Our results showed that TRIM56 knockout severely undermined the upregulation of ISGs by extracellular dsRNA and that loss of TRIM56 weakened the response to cytosolic dsDNA. ISG induction and ISGylation following IFN-α stimulation, however, were not compromised by TRIM56 deletion. Using a vesicular stomatitis virus-based antiviral bioactivity assay, we demonstrated that IFN-α could efficiently establish an antiviral state in TRIM56 null cells, providing direct evidence that TRIM56 is not required for the general antiviral action of IFN-I. Altogether, these data ascertain the contributions of TRIM56 to TLR3- and cGAS-STING-dependent antiviral pathways in HeLa cells and add to our understanding of the roles this protein plays in innate immunity.


Subject(s)
DNA/immunology , Interferon-alpha/immunology , RNA, Double-Stranded/immunology , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viruses/immunology , Animals , Chlorocebus aethiops , Cytosol/metabolism , HeLa Cells , Humans , Immunity, Innate , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Toll-Like Receptor 3/metabolism , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Vero Cells , Vesiculovirus/immunology
9.
PLoS Pathog ; 18(1): e1010250, 2022 01.
Article in English | MEDLINE | ID: mdl-35041722

ABSTRACT

ADARs (adenosine deaminases acting on RNA) are known for their adenosine-to-inosine RNA editing activity, and most recently, for their role in preventing aberrant dsRNA-response by activation of dsRNA sensors (i.e., RIG-I-like receptor homologs). However, it is still unclear whether suppressing spurious dsRNA-response represents the ancestral role of ADARs in bilaterians. As a first step to address this question, we identified ADAR1 and ADAR2 homologs in the planarian Schmidtea mediterranea, which is evolutionarily distant from canonical lab models (e.g., flies and nematodes). Our results indicate that knockdown of either planarian adar1 or adar2 by RNA interference (RNAi) resulted in upregulation of dsRNA-response genes, including three planarian rig-I-like receptor (prlr) homologs. Furthermore, independent knockdown of adar1 and adar2 reduced the number of infected cells with a dsRNA virus, suggesting they suppress a bona fide anti-viral dsRNA-response activity. Knockdown of adar1 also resulted in lesion formation and animal lethality, thus attesting to its essentiality. Simultaneous knockdown of adar1 and prlr1 rescued adar1(RNAi)-dependent animal lethality and rescued the dsRNA-response, suggesting that it contributes to the deleterious effect of adar1 knockdown. Finally, we found that ADAR2, but not ADAR1, mediates mRNA editing in planarians, suggesting at least in part non-redundant activities for planarians ADARs. Our results underline the essential role of ADARs in suppressing activation of harmful dsRNA-response in planarians, thus supporting it as their ancestral role in bilaterians. Our work also set the stage to study further and better understand the regulatory mechanisms governing anti-viral dsRNA-responses from an evolutionary standpoint using planarians as a model.


Subject(s)
Adenosine Deaminase/immunology , Adenosine Deaminase/metabolism , Planarians/physiology , RNA, Double-Stranded/immunology , Animals , Virus Diseases/immunology
10.
Signal Transduct Target Ther ; 7(1): 22, 2022 01 24.
Article in English | MEDLINE | ID: mdl-35075101

ABSTRACT

As a highly pathogenic human coronavirus, SARS-CoV-2 has to counteract an intricate network of antiviral host responses to establish infection and spread. The nucleic acid-induced stress response is an essential component of antiviral defense and is closely related to antiviral innate immunity. However, whether SARS-CoV-2 regulates the stress response pathway to achieve immune evasion remains elusive. In this study, SARS-CoV-2 NSP5 and N protein were found to attenuate antiviral stress granule (avSG) formation. Moreover, NSP5 and N suppressed IFN expression induced by infection of Sendai virus or transfection of a synthetic mimic of dsRNA, poly (I:C), inhibiting TBK1 and IRF3 phosphorylation, and restraining the nuclear translocalization of IRF3. Furthermore, HEK293T cells with ectopic expression of NSP5 or N protein were less resistant to vesicular stomatitis virus infection. Mechanistically, NSP5 suppressed avSG formation and disrupted RIG-I-MAVS complex to attenuate the RIG-I-mediated antiviral immunity. In contrast to the multiple targets of NSP5, the N protein specifically targeted cofactors upstream of RIG-I. The N protein interacted with G3BP1 to prevent avSG formation and to keep the cofactors G3BP1 and PACT from activating RIG-I. Additionally, the N protein also affected the recognition of dsRNA by RIG-I. This study revealed the intimate correlation between SARS-CoV-2, the stress response, and innate antiviral immunity, shedding light on the pathogenic mechanism of COVID-19.


Subject(s)
Coronavirus 3C Proteases/genetics , Coronavirus Nucleocapsid Proteins/genetics , DEAD Box Protein 58/genetics , DNA Helicases/genetics , Poly-ADP-Ribose Binding Proteins/genetics , RNA Helicases/genetics , RNA Recognition Motif Proteins/genetics , RNA-Binding Proteins/genetics , Receptors, Immunologic/genetics , SARS-CoV-2/genetics , Stress Granules/genetics , Animals , Chlorocebus aethiops , Coronavirus 3C Proteases/immunology , Coronavirus Nucleocapsid Proteins/immunology , DEAD Box Protein 58/immunology , DNA Helicases/immunology , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Immune Evasion , Phosphoproteins/genetics , Phosphoproteins/immunology , Poly I-C/pharmacology , Poly-ADP-Ribose Binding Proteins/immunology , Protein Binding , RNA Helicases/immunology , RNA Recognition Motif Proteins/immunology , RNA, Double-Stranded/genetics , RNA, Double-Stranded/immunology , RNA-Binding Proteins/immunology , Receptors, Immunologic/immunology , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Sendai virus/genetics , Sendai virus/immunology , Signal Transduction , Stress Granules/drug effects , Stress Granules/immunology , Stress Granules/virology , Vero Cells , Vesiculovirus/genetics , Vesiculovirus/immunology
11.
Fish Shellfish Immunol ; 121: 215-222, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34999226

ABSTRACT

Salmonids are one of the most farmed fish species worldwide. These aquatic vertebrates rely heavily on their innate immune responses as the first line of defense to defend themselves against invading pathogens. Although commercial vaccines are available against some viral and bacterial pathogens affecting salmonids, their protective efficacy varies. Using a prophylactic inducer of local and systemic innate immune responses to limit infection could have significant implications in salmonid aquaculture. A potent inducer of innate immune responses in fish is double-stranded RNA (dsRNA), a molecule that all viruses make during their replicative cycle. Polyinosinic: polycytidylic acid (polyI:C) is a synthetic dsRNA commonly used to induce type I interferons (IFNs), interferon stimulated genes (ISGs) as well as an antiviral state in vertebrate species. Based on in vitro data it was hypothesized that both local and systemic innate immune responses, in salmonids, would be enhanced by orally delivering high molecular weight polyI:C (HMW polyI:C) using cationic phytoglycogen nanoparticles (NPs) as a delivery method. The present study investigates this hypothesis using two feed delivery methods. In the first in vivo study, to ensure an equal distribution of dose, individual rainbow trout (Oncorhynchus mykiss) were orally gavaged with feed moistened with a solution containing HMW-NP (polyI:C complexed with cationic phytoglycogen nanoparticles) or HMW polyI:C alone. In a second in vivo experiment, to better mimic a more realistic feeding scenario, rainbow trout were fed feed pellets to which HMW, or HMW-NP was added. The expression of IFN1 and ISGs (vig-3, Mx1) were quantified using real-time PCR in the intestine (local response) and head kidney (systemic response). The results of these studies indicate that HMW-NP induced a higher level of IFN1 and ISG expression in the intestine and head kidney compared to the HMW fed fish. The results of this study could lead to new advances in therapeutics for the aquaculture industry by utilizing the innate immune response against invading pathogens using an orally delivered stimulant.


Subject(s)
Immunity, Innate , Interferon Type I , Nanoparticles , Oncorhynchus mykiss , RNA, Double-Stranded/immunology , Animals , Fish Diseases/prevention & control , Interferon Type I/immunology , Oncorhynchus mykiss/immunology
12.
Nucleic Acids Res ; 49(18): 10604-10617, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34520542

ABSTRACT

RNA hydrolysis presents problems in manufacturing, long-term storage, world-wide delivery and in vivo stability of messenger RNA (mRNA)-based vaccines and therapeutics. A largely unexplored strategy to reduce mRNA hydrolysis is to redesign RNAs to form double-stranded regions, which are protected from in-line cleavage and enzymatic degradation, while coding for the same proteins. The amount of stabilization that this strategy can deliver and the most effective algorithmic approach to achieve stabilization remain poorly understood. Here, we present simple calculations for estimating RNA stability against hydrolysis, and a model that links the average unpaired probability of an mRNA, or AUP, to its overall hydrolysis rate. To characterize the stabilization achievable through structure design, we compare AUP optimization by conventional mRNA design methods to results from more computationally sophisticated algorithms and crowdsourcing through the OpenVaccine challenge on the Eterna platform. We find that rational design on Eterna and the more sophisticated algorithms lead to constructs with low AUP, which we term 'superfolder' mRNAs. These designs exhibit a wide diversity of sequence and structure features that may be desirable for translation, biophysical size, and immunogenicity. Furthermore, their folding is robust to temperature, computer modeling method, choice of flanking untranslated regions, and changes in target protein sequence, as illustrated by rapid redesign of superfolder mRNAs for B.1.351, P.1 and B.1.1.7 variants of the prefusion-stabilized SARS-CoV-2 spike protein. Increases in in vitro mRNA half-life by at least two-fold appear immediately achievable.


Subject(s)
Algorithms , RNA, Double-Stranded/chemistry , RNA, Messenger/chemistry , RNA, Viral/chemistry , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Base Pairing , Base Sequence , COVID-19/prevention & control , Humans , Hydrolysis , RNA Stability , RNA, Double-Stranded/genetics , RNA, Double-Stranded/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Viral/genetics , RNA, Viral/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Thermodynamics
13.
J Gen Virol ; 102(7)2021 07.
Article in English | MEDLINE | ID: mdl-34236957

ABSTRACT

Mosquito-borne flaviviruses are significant contributors to the arboviral disease burdens both in Australia and globally. While routine arbovirus surveillance remains a vital exercise to identify known flaviviruses in mosquito populations, novel or divergent and emerging species can be missed by these traditional methods. The MAVRIC (monoclonal antibodies to viral RNA intermediates in cells) system is an ELISA-based method for broad-spectrum isolation of positive-sense and double-stranded RNA (dsRNA) viruses based on detection of dsRNA in infected cells. While the MAVRIC ELISA has successfully been used to detect known and novel flaviviruses in Australian mosquitoes, we previously reported that dsRNA could not be detected in dengue virus-infected cells using this method. In this study we identified additional flaviviruses which evade detection of dsRNA by the MAVRIC ELISA. Utilising chimeric flaviviruses we demonstrated that this outcome may be dictated by the non-structural proteins and/or untranslated regions of the flaviviral genome. In addition, we report a modified fixation method that enables improved detection of flavivirus dsRNA and inactivation of non-enveloped viruses from mosquito populations using the MAVRIC system. This study demonstrates the utility of anti-dsRNA monoclonal antibodies for identifying viral replication in insect and vertebrate cell systems and highlights a unique characteristic of flavivirus replication.


Subject(s)
Culicidae/virology , Flavivirus/isolation & purification , Flavivirus/physiology , RNA, Double-Stranded/analysis , RNA, Viral/analysis , Aedes/virology , Animals , Antibodies, Monoclonal , Australia , Cell Line , Dengue Virus/genetics , Dengue Virus/isolation & purification , Dengue Virus/physiology , Enzyme-Linked Immunosorbent Assay , Flavivirus/genetics , RNA, Double-Stranded/immunology , RNA, Viral/immunology , Viral Envelope Proteins/analysis , Viral Envelope Proteins/metabolism , Viral Nonstructural Proteins/analysis , Viral Nonstructural Proteins/metabolism , Virus Replication
14.
Nature ; 597(7874): 109-113, 2021 09.
Article in English | MEDLINE | ID: mdl-34261127

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) is a cytosolic DNA sensor that produces the second messenger cG[2'-5']pA[3'-5']p (2'3'-cGAMP) and controls activation of innate immunity in mammalian cells1-5. Animal genomes typically encode multiple proteins with predicted homology to cGAS6-10, but the function of these uncharacterized enzymes is unknown. Here we show that cGAS-like receptors (cGLRs) are innate immune sensors that are capable of recognizing divergent molecular patterns and catalysing synthesis of distinct nucleotide second messenger signals. Crystal structures of human and insect cGLRs reveal a nucleotidyltransferase signalling core shared with cGAS and a diversified primary ligand-binding surface modified with notable insertions and deletions. We demonstrate that surface remodelling of cGLRs enables altered ligand specificity and used a forward biochemical screen to identify cGLR1 as a double-stranded RNA sensor in the model organism Drosophila melanogaster. We show that RNA recognition activates Drosophila cGLR1 to synthesize the novel product cG[3'-5']pA[2'-5']p (3'2'-cGAMP). A crystal structure of Drosophila stimulator of interferon genes (dSTING) in complex with 3'2'-cGAMP explains selective isomer recognition, and 3'2'-cGAMP induces an enhanced antiviral state in vivo that protects from viral infection. Similar to radiation of Toll-like receptors in pathogen immunity, our results establish cGLRs as a diverse family of metazoan pattern recognition receptors.


Subject(s)
Drosophila melanogaster/metabolism , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/metabolism , RNA, Double-Stranded/metabolism , Receptors, Pattern Recognition/metabolism , Second Messenger Systems , Amino Acid Sequence , Animals , Crystallography, X-Ray , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Drosophila melanogaster/virology , Female , Humans , Immunity, Innate , Male , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/immunology , RNA, Double-Stranded/analysis , RNA, Double-Stranded/immunology , Receptors, Pattern Recognition/chemistry , Receptors, Pattern Recognition/immunology , Viruses/immunology
15.
Nature ; 597(7874): 114-118, 2021 09.
Article in English | MEDLINE | ID: mdl-34261128

ABSTRACT

In mammals, cyclic GMP-AMP (cGAMP) synthase (cGAS) produces the cyclic dinucleotide 2'3'-cGAMP in response to cytosolic DNA and this triggers an antiviral immune response. cGAS belongs to a large family of cGAS/DncV-like nucleotidyltransferases that is present in both prokaryotes1 and eukaryotes2-5. In bacteria, these enzymes synthesize a range of cyclic oligonucleotides and have recently emerged as important regulators of phage infections6-8. Here we identify two cGAS-like receptors (cGLRs) in the insect Drosophila melanogaster. We show that cGLR1 and cGLR2 activate Sting- and NF-κB-dependent antiviral immunity in response to infection with RNA or DNA viruses. cGLR1 is activated by double-stranded RNA to produce the cyclic dinucleotide 3'2'-cGAMP, whereas cGLR2 produces a combination of 2'3'-cGAMP and 3'2'-cGAMP in response to an as-yet-unidentified stimulus. Our data establish cGAS as the founding member of a family of receptors that sense different types of nucleic acids and trigger immunity through the production of cyclic dinucleotides beyond 2'3'-cGAMP.


Subject(s)
Drosophila melanogaster/immunology , Nucleotidyltransferases/immunology , Receptors, Pattern Recognition/metabolism , Viruses/immunology , Amino Acid Sequence , Animals , Cell Line , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/virology , Female , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Ligands , Male , Membrane Proteins/metabolism , Models, Molecular , NF-kappa B/metabolism , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/classification , Nucleotidyltransferases/deficiency , Nucleotidyltransferases/metabolism , RNA, Double-Stranded/analysis , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , Receptors, Pattern Recognition/classification , Receptors, Pattern Recognition/deficiency , Receptors, Pattern Recognition/immunology
16.
Cell Rep ; 35(2): 108976, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852834

ABSTRACT

RIG-I-like receptors (RLRs) are involved in the discrimination of self versus non-self via the recognition of double-stranded RNA (dsRNA). Emerging evidence suggests that immunostimulatory dsRNAs are ubiquitously expressed but are disrupted or sequestered by cellular RNA binding proteins (RBPs). TDP-43 is an RBP associated with multiple neurological disorders and is essential for cell viability. Here, we demonstrate that TDP-43 regulates the accumulation of immunostimulatory dsRNA. The immunostimulatory RNA is identified as RNA polymerase III transcripts, including 7SL and Alu retrotransposons, and we demonstrate that the RNA-binding activity of TDP-43 is required to prevent immune stimulation. The dsRNAs activate a RIG-I-dependent interferon (IFN) response, which promotes necroptosis. Genetic inactivation of the RLR-pathway rescues the interferon-mediated cell death associated with loss of TDP-43. Collectively, our study describes a role for TDP-43 in preventing the accumulation of endogenous immunostimulatory dsRNAs and uncovers an intricate relationship between the control of cellular gene expression and IFN-mediated cell death.


Subject(s)
DEAD Box Protein 58/genetics , DNA-Binding Proteins/genetics , Herpesvirus 8, Human/genetics , Necroptosis/genetics , RNA, Double-Stranded/genetics , Receptors, Immunologic/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Alu Elements , Cell Line, Tumor , Cell Survival , Cytokines/genetics , Cytokines/immunology , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/immunology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/immunology , Epithelial Cells/immunology , Epithelial Cells/virology , Gene Expression Regulation , HEK293 Cells , Herpesvirus 8, Human/growth & development , Herpesvirus 8, Human/immunology , Humans , Immunization , Interferons/genetics , Interferons/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Necroptosis/immunology , Neurons/immunology , Neurons/virology , RNA Polymerase III/genetics , RNA Polymerase III/immunology , RNA, Double-Stranded/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA, Small Cytoplasmic/genetics , RNA, Small Cytoplasmic/immunology , RNA, Viral/genetics , RNA, Viral/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/immunology , Signal Recognition Particle/genetics , Signal Recognition Particle/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Ubiquitins/genetics , Ubiquitins/immunology
17.
Genes (Basel) ; 13(1)2021 12 23.
Article in English | MEDLINE | ID: mdl-35052382

ABSTRACT

ADAR1-mediated deamination of adenosines in long double-stranded RNAs plays an important role in modulating the innate immune response. However, recent investigations based on metatranscriptomic samples of COVID-19 patients and SARS-COV-2-infected Vero cells have recovered contrasting findings. Using RNAseq data from time course experiments of infected human cell lines and transcriptome data from Vero cells and clinical samples, we prove that A-to-G changes observed in SARS-COV-2 genomes represent genuine RNA editing events, likely mediated by ADAR1. While the A-to-I editing rate is generally low, changes are distributed along the entire viral genome, are overrepresented in exonic regions, and are (in the majority of cases) nonsynonymous. The impact of RNA editing on virus-host interactions could be relevant to identify potential targets for therapeutic interventions.


Subject(s)
Adenosine Deaminase/genetics , COVID-19/genetics , Genome, Viral , Host-Pathogen Interactions/genetics , RNA Editing , RNA, Viral/genetics , RNA-Binding Proteins/genetics , SARS-CoV-2/genetics , Adenosine/metabolism , Adenosine Deaminase/immunology , Animals , COVID-19/metabolism , COVID-19/virology , Cell Line, Tumor , Chlorocebus aethiops , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Deamination , Epithelial Cells/immunology , Epithelial Cells/virology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Inosine/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Interferon-Induced Helicase, IFIH1/immunology , Interferon-beta/genetics , Interferon-beta/immunology , RNA, Double-Stranded/genetics , RNA, Double-Stranded/immunology , RNA, Viral/immunology , RNA-Binding Proteins/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Transcriptome , Vero Cells
18.
Dev Comp Immunol ; 116: 103931, 2021 03.
Article in English | MEDLINE | ID: mdl-33220355

ABSTRACT

The enzyme Dicer is best known for its role as an endoribonuclease in the small RNA pathway, playing a crucial role in recognizing viral double-stranded RNA (dsRNA) and inducing down-stream cascades to mediate anti-virus immunity. In the present study, a truncated Dicer-like gene was identified from oyster Crassostrea gigas, and its open reading frame (ORF) encoded a polypeptide (designed as CgDCL) of 530 amino acids. The CgDCL contained one N-terminal DEAD domain and a C-terminal helicase domain, but lack the conserved PAZ domain, ribonuclease domain (RIBOc) and dsRNA binding domain. The mRNA transcripts of CgDCL were detected in all the examined tissues with high expression levels in lip, gills and haemocytes, which were 62.06-fold, 48.91-fold and 47.13-fold (p < 0.05) of that in mantle, respectively. In the primarily cultured oyster haemocytes, the mRNA transcripts of CgDCL were significantly induced at 12 h after poly(I:C) stimulation, which were 4.04-fold (p < 0.05) of that in control group. The expression level of CgDCL mRNA in haemocytes was up-regulated significantly after dsRNA and recombinant interferon-like protein (rCgIFNLP) injection, which was 12.87-fold (p < 0.01) and 3.22-fold (p < 0.05) of that in control group, respectively. CgDCL proteins were mainly distributed in the cytoplasm of haemocytes. The recombinant CgDCL protein displayed binding activity to dsRNA and poly(I:C), but no obvious dsRNA cleavage activity. These results collectively suggest that truncated CgDCL from C. gigas was able to be activated by poly(I:C), dsRNA and CgIFNLP, and functioned as an intracellular recognition molecule to bind nucleic acid of virus, indicating a potential mutual cooperation between RNAi and IFN-like system in anti-virus immunity of oysters.


Subject(s)
Crassostrea/immunology , Immunity, Innate , Ribonuclease III/immunology , Amino Acid Sequence , Animals , Cytoplasm/metabolism , Hemocytes/immunology , Hemocytes/metabolism , Interferons/immunology , Interferons/metabolism , Open Reading Frames , Phylogeny , Poly I-C/immunology , Poly I-C/metabolism , Protein Domains , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Sequence Alignment , Signal Transduction/immunology
19.
J Immunol ; 205(12): 3419-3428, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33188074

ABSTRACT

Melanoma differentiation-associated gene 5 (MDA5) is a key cytoplasmic dsRNA sensor. Upon binding to invading viral RNA, activated MDA5 is recruited to mitochondria and interacts with mitochondrial antiviral signaling gene (MAVS) to initiate innate antiviral immune responses. The elegant regulation of this process remains elusive. In this study, using the Chinese tree shrew (Tupaia belangeri chinensis), which is genetically close to primates, we identified the Tupaia oligoadenylate synthetases-like 1 (tOASL1) as a positive regulator of the Tupaia MDA5 (tMDA5) and Tupaia MAVS (tMAVS)-mediated IFN signaling. Overexpression of tOASL1 significantly potentiated the RNA virus-triggered induction of the type I IFNs and downstream antiviral genes. Conversely, knockdown of tOASL1 had an impaired antiviral immune response. Mechanistically, tOASL1 was associated with mitochondria and directly interacted with tMDA5 and tMAVS. Upon RNA virus infection, tOASL1 enhanced the interaction between tMDA5 and tMAVS via its OAS and UBL domains. Our results revealed a novel mechanism by which tOASL1 contributes to host antiviral responses via enhancing tMDA5 and tMAVS interaction.


Subject(s)
2',5'-Oligoadenylate Synthetase/immunology , Adaptor Proteins, Signal Transducing/immunology , Immunity, Innate , Interferon-Induced Helicase, IFIH1/immunology , RNA Virus Infections/immunology , RNA Viruses/immunology , RNA, Double-Stranded/immunology , RNA, Viral/immunology , Animals , Tupaia
20.
J Immunol ; 205(12): 3408-3418, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33177158

ABSTRACT

The RIG-I receptor induces the innate antiviral responses upon sensing RNA viruses. The mechanisms through which RIG-I optimizes the strength of the downstream signaling remain incompletely understood. In this study, we identified that NSUN5 could potentiate the RIG-I innate signaling pathway. Deficiency of NSUN5 enhanced RNA virus proliferation and inhibited the induction of the downstream antiviral genes. Consistently, NSUN5-deficient mice were more susceptible to RNA virus infection than their wild-type littermates. Mechanistically, NSUN5 bound directly to both viral RNA and RIG-I, synergizing the recognition of dsRNA by RIG-I. Collectively, to our knowledge, this study characterized NSUN5 as a novel RIG-I coreceptor, playing a vital role in restricting RNA virus infection.


Subject(s)
DEAD Box Protein 58/immunology , Methyltransferases/immunology , Muscle Proteins/immunology , RNA Virus Infections/immunology , RNA Viruses/immunology , RNA, Double-Stranded/immunology , RNA, Viral/immunology , Receptors, Immunologic/immunology , tRNA Methyltransferases/immunology , Animals , Chlorocebus aethiops , HEK293 Cells , Humans , Immunity, Innate , Mice , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...