1
|
Sharma D, Chakraborty S. RNA editing sites and triplet usage in exomes of bat RNA virus genomes of the family Paramyxoviridae. Microb Pathog 2024; 194:106796. [PMID: 39025379 DOI: 10.1016/j.micpath.2024.106796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Bats contain a diverse spectrum of viral species in their bodies. The RNA virus family Paramyxoviridae tends to infect several vertebrate species, which are accountable for a variety of devastating infections in both humans and animals. Viruses of this kind include measles, mumps, and Hendra. Some synonymous codons are favoured over others in mRNAs during gene-to-protein synthesis process. Such phenomenon is termed as codon usage bias (CUB). Our research emphasized many aspects that shape the CUB of genes in the Paramyxoviridae family found in bats. Here, the nitrogenous base A occurred the most. AT was found to be abundant in the coding sequences of the Paramyxoviridae family. RSCU data revealed that A or T ending codons occurred more frequently than predicted. Furthermore, 3 overrepresented codons (CAT, AGA, and GCA) and 7 underrepresented codons (CCG, TCG, CGC, CGG, CGT, GCG and ACG) were detected in the viral genomes. Correspondence analysis, neutrality plot, and parity plots highlight the combined impact of mutational pressure and natural selection on CUB. The neutrality plot of GC12 against GC3 yielded a regression coefficient value of 0.366, indicating that natural selection had a significant (63.4 %) impact. Moreover, RNA editing analysis was done, which revealed the highest frequency of C to T mutations. The results of our research revealed the pattern of codon usage and RNA editing sites in Paramyxoviridae genomes.
Collapse
Affiliation(s)
- Deepika Sharma
- Department of Biotechnology, Assam University, Silchar, 788011, Assam, India.
| | - Supriyo Chakraborty
- Department of Biotechnology, Assam University, Silchar, 788011, Assam, India.
| |
Collapse
|
2
|
Zheng YY, Reddy K, Vangaveti S, Sheng J. Inosine-Induced Base Pairing Diversity during Reverse Transcription. ACS Chem Biol 2024; 19:348-356. [PMID: 38252964 PMCID: PMC10877575 DOI: 10.1021/acschembio.3c00555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/27/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024]
Abstract
A-to-I editing catalyzed by adenosine deaminase acting on RNAs impacts numerous physiological and biochemical processes that are essential for cellular functions and is a big contributor to the infectivity of certain RNA viruses. The outcome of this deamination leads to changes in the eukaryotic transcriptome functionally resembling A-G transitions since inosine preferentially pairs with cytosine. Moreover, hyper-editing or multiple A to G transitions in clusters were detected in measles virus. Inosine modifications either directly on viral RNA or on cellular RNA can have antiviral or pro-viral repercussions. While many of the significant roles of inosine in cellular RNAs are well understood, the effects of hyper-editing of A to I on viral polymerase activity during RNA replication remain elusive. Moreover, biological strategies such as molecular cloning and RNA-seq for transcriptomic interrogation rely on RT-polymerase chain reaction with little to no emphasis placed on the first step, reverse transcription, which may reshape the sequencing results when hypermodification is present. In this study, we systematically explore the influence of inosine modification, varying the number and position of inosines, on decoding outcomes using three different reverse transcriptases (RTs) followed by standard Sanger sequencing. We find that inosine alone or in clusters can differentially affect the RT activity. To gain structural insights into the accommodation of inosine in the polymerase site of HIV-1 reverse transcriptase (HIV-1-RT) and how this structural context affects the base pairing rules for inosine, we performed molecular dynamics simulations of the HIV-1-RT. The simulations highlight the importance of the protein-nucleotide interaction as a critical factor in deciphering the base pairing behavior of inosine clusters. This effort sets the groundwork for decrypting the physiological significance of inosine and linking the fidelity of reverse transcriptase and the possible diverse transcription outcomes of cellular RNAs and/or viral RNAs where hyper-edited inosines are present in the transcripts.
Collapse
Affiliation(s)
- Ya Ying Zheng
- Department
of Chemistry, University at Albany, State
University of New York, 1400 Washington Avenue, Albany, New York 12222, United States
- The
RNA Institute, University at Albany, State
University of New York, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Kaalak Reddy
- The
RNA Institute, University at Albany, State
University of New York, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Sweta Vangaveti
- The
RNA Institute, University at Albany, State
University of New York, 1400 Washington Avenue, Albany, New York 12222, United States
| | - Jia Sheng
- Department
of Chemistry, University at Albany, State
University of New York, 1400 Washington Avenue, Albany, New York 12222, United States
- The
RNA Institute, University at Albany, State
University of New York, 1400 Washington Avenue, Albany, New York 12222, United States
| |
Collapse
|
3
|
Roy A, Ghosh A. Epigenetic Restriction Factors (eRFs) in Virus Infection. Viruses 2024; 16:183. [PMID: 38399958 PMCID: PMC10892949 DOI: 10.3390/v16020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
The ongoing arms race between viruses and their hosts is constantly evolving. One of the ways in which cells defend themselves against invading viruses is by using restriction factors (RFs), which are cell-intrinsic antiviral mechanisms that block viral replication and transcription. Recent research has identified a specific group of RFs that belong to the cellular epigenetic machinery and are able to restrict the gene expression of certain viruses. These RFs can be referred to as epigenetic restriction factors or eRFs. In this review, eRFs have been classified into two categories. The first category includes eRFs that target viral chromatin. So far, the identified eRFs in this category include the PML-NBs, the KRAB/KAP1 complex, IFI16, and the HUSH complex. The second category includes eRFs that target viral RNA or, more specifically, the viral epitranscriptome. These epitranscriptomic eRFs have been further classified into two types: those that edit RNA bases-adenosine deaminase acting on RNA (ADAR) and pseudouridine synthases (PUS), and those that covalently modify viral RNA-the N6-methyladenosine (m6A) writers, readers, and erasers. We delve into the molecular machinery of eRFs, their role in limiting various viruses, and the mechanisms by which viruses have evolved to counteract them. We also examine the crosstalk between different eRFs, including the common effectors that connect them. Finally, we explore the potential for new discoveries in the realm of epigenetic networks that restrict viral gene expression, as well as the future research directions in this area.
Collapse
Affiliation(s)
- Arunava Roy
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA;
| | | |
Collapse
|
4
|
Zhu T, Niu G, Zhang Y, Chen M, Li CY, Hao L, Zhang Z. Host-mediated RNA editing in viruses. Biol Direct 2023; 18:12. [PMID: 36978112 PMCID: PMC10043548 DOI: 10.1186/s13062-023-00366-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Viruses rely on hosts for life and reproduction, cause a variety of symptoms from common cold to AIDS to COVID-19 and provoke public health threats claiming millions of lives around the globe. RNA editing, as a crucial co-/post-transcriptional modification inducing nucleotide alterations on both endogenous and exogenous RNA sequences, exerts significant influences on virus replication, protein synthesis, infectivity and toxicity. Hitherto, a number of host-mediated RNA editing sites have been identified in diverse viruses, yet lacking a full picture of RNA editing-associated mechanisms and effects in different classes of viruses. Here we synthesize the current knowledge of host-mediated RNA editing in a variety of viruses by considering two enzyme families, viz., ADARs and APOBECs, thereby presenting a landscape of diverse editing mechanisms and effects between viruses and hosts. In the ongoing pandemic, our study promises to provide potentially valuable insights for better understanding host-mediated RNA editing on ever-reported and newly-emerging viruses.
Collapse
Affiliation(s)
- Tongtong Zhu
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guangyi Niu
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuansheng Zhang
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ming Chen
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chuan-Yun Li
- Laboratory of Bioinformatics and Genomic Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Lili Hao
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
- China National Center for Bioinformation, Beijing, 100101, China.
| | - Zhang Zhang
- National Genomics Data Center & CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
- China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
5
|
Martignano F, Di Giorgio S, Mattiuz G, Conticello SG. Commentary on "Poor evidence for host-dependent regular RNA editing in the transcriptome of SARS-CoV-2". J Appl Genet 2022; 63:423-428. [PMID: 35279801 PMCID: PMC8917825 DOI: 10.1007/s13353-022-00688-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/14/2022] [Accepted: 02/16/2022] [Indexed: 01/10/2023]
Abstract
Analysis of the SARS-CoV-2 transcriptome has revealed a background of low-frequency intra-host genetic changes with a strong bias towards transitions. A similar pattern is also observed when inter-host variability is considered. We and others have shown that the cellular RNA editing machinery based on ADAR and APOBEC host-deaminases could be involved in the onset of SARS-CoV-2 genetic variability. Our hypothesis is based both on similarities with other known forms of viral genome editing and on the excess of transition changes, which is difficult to explain with errors during viral replication. Zong et al. criticize our analysis on both conceptual and technical grounds. While ultimate proof of an involvement of host deaminases in viral RNA editing will depend on experimental validation, here, we address the criticism to suggest that viral RNA editing is the most reasonable explanation for the observed intra- and inter-host variability.
Collapse
Affiliation(s)
- F Martignano
- Core Research Laboratory, ISPRO, 50139, Firenze, Italy
| | - S Di Giorgio
- German Cancer Research Center (DKFZ), Division of Immune Diversity, Foundation Under Public Law, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - G Mattiuz
- Department of Experimental and Clinical Medicine, University of Florence, 50139, Firenze, Italy
| | - S G Conticello
- Core Research Laboratory, ISPRO, 50139, Firenze, Italy.
- Institute of Clinical Physiology, National Research Council, 56124, Pisa, Italy.
| |
Collapse
|
6
|
Dutta N, Deb I, Sarzynska J, Lahiri A. Inosine and its methyl derivatives: Occurrence, biogenesis, and function in RNA. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 169-170:21-52. [PMID: 35065168 DOI: 10.1016/j.pbiomolbio.2022.01.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/11/2021] [Accepted: 01/11/2022] [Indexed: 05/21/2023]
Abstract
Inosine is one of the most common post-transcriptional modifications. Since its discovery, it has been noted for its ability to contribute to non-Watson-Crick interactions within RNA. Rapidly accumulating evidence points to the widespread generation of inosine through hydrolytic deamination of adenosine to inosine by different classes of adenosine deaminases. Three naturally occurring methyl derivatives of inosine, i.e., 1-methylinosine, 2'-O-methylinosine and 1,2'-O-dimethylinosine are currently reported in RNA modification databases. These modifications are expected to lead to changes in the structure, folding, dynamics, stability and functions of RNA. The importance of the modifications is indicated by the strong conservation of the modifying enzymes across organisms. The structure, binding and catalytic mechanism of the adenosine deaminases have been well-studied, but the underlying mechanism of the catalytic reaction is not very clear yet. Here we extensively review the existing data on the occurrence, biogenesis and functions of inosine and its methyl derivatives in RNA. We also included the structural and thermodynamic aspects of these modifications in our review to provide a detailed and integrated discussion on the consequences of A-to-I editing in RNA and the contribution of different structural and thermodynamic studies in understanding its role in RNA. We also highlight the importance of further studies for a better understanding of the mechanisms of the different classes of deamination reactions. Further investigation of the structural and thermodynamic consequences and functions of these modifications in RNA should provide more useful information about their role in different diseases.
Collapse
Affiliation(s)
- Nivedita Dutta
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92, Acharya Prafulla Chandra Road, Kolkata, 700009, West Bengal, India
| | - Indrajit Deb
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92, Acharya Prafulla Chandra Road, Kolkata, 700009, West Bengal, India
| | - Joanna Sarzynska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704, Poznan, Poland
| | - Ansuman Lahiri
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92, Acharya Prafulla Chandra Road, Kolkata, 700009, West Bengal, India.
| |
Collapse
|
7
|
Impact of ADAR-induced editing of minor viral RNA populations on replication and transmission of SARS-CoV-2. Proc Natl Acad Sci U S A 2022; 119:2112663119. [PMID: 35064076 PMCID: PMC8833170 DOI: 10.1073/pnas.2112663119] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2021] [Indexed: 12/19/2022] Open
Abstract
Viral RNA may be edited by enzymes of the ADAR family that deaminate adenosine residues with ensuing A→G mutations. We found multiple A→G mutations in minor viral populations of the SARS-CoV-2 genome. A→G mutations accumulated in the receptor binding domain of the spike gene, which may cause structural changes by altering binding to the ACE2 receptor. Presence of A→G mutations in minor viral populations was associated with reduced viral load, implying that ADAR may limit viral replication. Analyses of >250,000 European samples from 2020 revealed that A→G mutations in SARS-CoV-2 RNA were inversely correlated with mortality as a reflection of incidence. ADAR may thus be important in providing new variants of SARS-CoV-2 with altered infectivity and transmissibility. Adenosine deaminases acting on RNA (ADAR) are RNA-editing enzymes that may restrict viral infection. We have utilized deep sequencing to determine adenosine to guanine (A→G) mutations, signifying ADAR activity, in clinical samples retrieved from 93 severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)–infected patients in the early phase of the COVID-19 pandemic. A→G mutations were detected in 0.035% (median) of RNA residues and were predominantly nonsynonymous. These mutations were rarely detected in the major viral population but were abundant in minor viral populations in which A→G was more prevalent than any other mutation (P < 0.001). The A→G substitutions accumulated in the spike protein gene at positions corresponding to amino acids 505 to 510 in the receptor binding motif and at amino acids 650 to 655. The frequency of A→G mutations in minor viral populations was significantly associated with low viral load (P < 0.001). We additionally analyzed A→G mutations in 288,247 SARS-CoV-2 major (consensus) sequences representing the dominant viral population. The A→G mutations observed in minor viral populations in the initial patient cohort were increasingly detected in European consensus sequences between March and June 2020 (P < 0.001) followed by a decline of these mutations in autumn and early winter (P < 0.001). We propose that ADAR-induced deamination of RNA is a significant source of mutated SARS-CoV-2 and hypothesize that the degree of RNA deamination may determine or reflect viral fitness and infectivity.
Collapse
|
8
|
Dual isoform sequencing reveals complex transcriptomic and epitranscriptomic landscapes of a prototype baculovirus. Sci Rep 2022; 12:1291. [PMID: 35079129 PMCID: PMC8789824 DOI: 10.1038/s41598-022-05457-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 01/05/2022] [Indexed: 12/23/2022] Open
Abstract
In this study, two long-read sequencing (LRS) techniques, MinION from Oxford Nanopore Technologies and Sequel from the Pacific Biosciences, were used for the transcriptional characterization of a prototype baculovirus, Autographa californica multiple nucleopolyhedrovirus. LRS is able to read full-length RNA molecules, and thereby distinguish between transcript isoforms, mono- and polycistronic RNAs, and overlapping transcripts. Altogether, we detected 875 transcript species, of which 759 were novel and 116 were annotated previously. These RNA molecules include 41 novel putative protein coding transcripts [each containing 5'-truncated in-frame open reading frames (ORFs), 14 monocistronic transcripts, 99 polygenic RNAs, 101 non-coding RNAs, and 504 untranslated region isoforms. This work also identified novel replication origin-associated transcripts, upstream ORFs, cis-regulatory sequences and poly(A) sites. We also detected RNA methylation in 99 viral genes and RNA hyper-editing in the longer 5'-UTR transcript isoform of the canonical ORF 19 transcript.
Collapse
|
9
|
Piontkivska H, Wales-McGrath B, Miyamoto M, Wayne ML. ADAR Editing in Viruses: An Evolutionary Force to Reckon with. Genome Biol Evol 2021; 13:evab240. [PMID: 34694399 PMCID: PMC8586724 DOI: 10.1093/gbe/evab240] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 02/06/2023] Open
Abstract
Adenosine Deaminases that Act on RNA (ADARs) are RNA editing enzymes that play a dynamic and nuanced role in regulating transcriptome and proteome diversity. This editing can be highly selective, affecting a specific site within a transcript, or nonselective, resulting in hyperediting. ADAR editing is important for regulating neural functions and autoimmunity, and has a key role in the innate immune response to viral infections, where editing can have a range of pro- or antiviral effects and can contribute to viral evolution. Here we examine the role of ADAR editing across a broad range of viral groups. We propose that the effect of ADAR editing on viral replication, whether pro- or antiviral, is better viewed as an axis rather than a binary, and that the specific position of a given virus on this axis is highly dependent on virus- and host-specific factors, and can change over the course of infection. However, more research needs to be devoted to understanding these dynamic factors and how they affect virus-ADAR interactions and viral evolution. Another area that warrants significant attention is the effect of virus-ADAR interactions on host-ADAR interactions, particularly in light of the crucial role of ADAR in regulating neural functions. Answering these questions will be essential to developing our understanding of the relationship between ADAR editing and viral infection. In turn, this will further our understanding of the effects of viruses such as SARS-CoV-2, as well as many others, and thereby influence our approach to treating these deadly diseases.
Collapse
Affiliation(s)
- Helen Piontkivska
- Department of Biological Sciences, Kent State University, Ohio, USA
- School of Biomedical Sciences, Kent State University, Ohio, USA
- Brain Health Research Institute, Kent State University, Ohio, USA
| | | | - Michael Miyamoto
- Department of Biology, University of Florida, Gainesville, Florida, USA
| | - Marta L Wayne
- Department of Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
10
|
Abstract
C6 deamination of adenosine (A) to inosine (I) in double-stranded RNA (dsRNA) is catalyzed by a family of enzymes known as ADARs (adenosine deaminases acting on RNA) encoded by three genes in mammals. Alternative promoters and splicing produce two ADAR1 proteins, an interferon-inducible cytoplasmic p150 and a constitutively expressed p110 that like ADAR2 is a nuclear enzyme. ADAR3 lacks deaminase activity. A-to-I editing occurs with both viral and cellular RNAs. Deamination activity is dependent on dsRNA substrate structure and regulatory RNA-binding proteins and ranges from highly site selective with hepatitis D RNA and glutamate receptor precursor messenger RNA (pre-mRNA) to hyperediting of measles virus and polyomavirus transcripts and cellular inverted Alu elements. Because I base-pairs as guanosine instead of A, editing can alter mRNA decoding, pre-mRNA splicing, and microRNA silencing. Editing also alters dsRNA structure, thereby suppressing innate immune responses including interferon production and action. Expected final online publication date for the Annual Review of Virology, Volume 8 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Christian K Pfaller
- Division of Veterinary Medicine, Paul-Ehrlich-Institute, Langen 63225, Germany
| | - Cyril X George
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, California 93106, USA;
| | - Charles E Samuel
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, California 93106, USA;
| |
Collapse
|
11
|
Srinivasan S, Torres AG, Ribas de Pouplana L. Inosine in Biology and Disease. Genes (Basel) 2021; 12:600. [PMID: 33921764 PMCID: PMC8072771 DOI: 10.3390/genes12040600] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023] Open
Abstract
The nucleoside inosine plays an important role in purine biosynthesis, gene translation, and modulation of the fate of RNAs. The editing of adenosine to inosine is a widespread post-transcriptional modification in transfer RNAs (tRNAs) and messenger RNAs (mRNAs). At the wobble position of tRNA anticodons, inosine profoundly modifies codon recognition, while in mRNA, inosines can modify the sequence of the translated polypeptide or modulate the stability, localization, and splicing of transcripts. Inosine is also found in non-coding and exogenous RNAs, where it plays key structural and functional roles. In addition, molecular inosine is an important secondary metabolite in purine metabolism that also acts as a molecular messenger in cell signaling pathways. Here, we review the functional roles of inosine in biology and their connections to human health.
Collapse
Affiliation(s)
- Sundaramoorthy Srinivasan
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, 08028 Barcelona, Catalonia, Spain; (S.S.); (A.G.T.)
| | - Adrian Gabriel Torres
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, 08028 Barcelona, Catalonia, Spain; (S.S.); (A.G.T.)
| | - Lluís Ribas de Pouplana
- Institute for Research in Biomedicine, Barcelona Institute of Science and Technology, 08028 Barcelona, Catalonia, Spain; (S.S.); (A.G.T.)
- Catalan Institution for Research and Advanced Studies, 08010 Barcelona, Catalonia, Spain
| |
Collapse
|
12
|
Kosuge M, Furusawa-Nishii E, Ito K, Saito Y, Ogasawara K. Point mutation bias in SARS-CoV-2 variants results in increased ability to stimulate inflammatory responses. Sci Rep 2020; 10:17766. [PMID: 33082451 PMCID: PMC7575582 DOI: 10.1038/s41598-020-74843-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/08/2020] [Indexed: 02/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection induces severe pneumonia and is the cause of a worldwide pandemic. Coronaviruses, including SARS-CoV-2, have RNA proofreading enzymes in their genomes, resulting in fewer gene mutations than other RNA viruses. Nevertheless, variants of SARS-CoV-2 exist and may induce different symptoms; however, the factors and the impacts of these mutations are not well understood. We found that there is a bias to the mutations occurring in SARS-CoV-2 variants, with disproportionate mutation to uracil (U). These point mutations to U are mainly derived from cytosine (C), which is consistent with the substrate specificity of host RNA editing enzymes, APOBECs. We also found the point mutations which are consistent with other RNA editing enzymes, ADARs. For the C-to-U mutations, the context of the upstream uracil and downstream guanine from mutated position was found to be most prevalent. Further, the degree of increase of U in SARS-CoV-2 variants correlates with enhanced production of cytokines, such as TNF-α and IL-6, in cell lines when compared with stimulation by the ssRNA sequence of the isolated virus in Wuhan. Therefore, RNA editing is a factor for mutation bias in SARS-CoV-2 variants, which affects host inflammatory cytokines production.
Collapse
Affiliation(s)
- Masato Kosuge
- Department of Immunobiology, Institute of Development Aging and Cancer, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Emi Furusawa-Nishii
- Department of Immunobiology, Institute of Development Aging and Cancer, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Koyu Ito
- Department of Immunobiology, Institute of Development Aging and Cancer, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Yoshiro Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Miyagi, Japan
| | - Kouetsu Ogasawara
- Department of Immunobiology, Institute of Development Aging and Cancer, 4-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| |
Collapse
|
13
|
Vogel OA, Han J, Liang CY, Manicassamy S, Perez JT, Manicassamy B. The p150 Isoform of ADAR1 Blocks Sustained RLR signaling and Apoptosis during Influenza Virus Infection. PLoS Pathog 2020; 16:e1008842. [PMID: 32898178 PMCID: PMC7500621 DOI: 10.1371/journal.ppat.1008842] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/18/2020] [Accepted: 07/28/2020] [Indexed: 12/24/2022] Open
Abstract
Signaling through retinoic acid inducible gene I (RIG-I) like receptors (RLRs) is tightly regulated, with activation occurring upon sensing of viral nucleic acids, and suppression mediated by negative regulators. Under homeostatic conditions aberrant activation of melanoma differentiation-associated protein-5 (MDA5) is prevented through editing of endogenous dsRNA by RNA editing enzyme Adenosine Deaminase Acting on RNA (ADAR1). In addition, ADAR1 is postulated to play pro-viral and antiviral roles during viral infections that are dependent or independent of RNA editing activity. Here, we investigated the importance of ADAR1 isoforms in modulating influenza A virus (IAV) replication and revealed the opposing roles for ADAR1 isoforms, with the nuclear p110 isoform restricting versus the cytoplasmic p150 isoform promoting IAV replication. Importantly, we demonstrate that p150 is critical for preventing sustained RIG-I signaling, as p150 deficient cells showed increased IFN-β expression and apoptosis during IAV infection, independent of RNA editing activity. Taken together, the p150 isoform of ADAR1 is important for preventing sustained RIG-I induced IFN-β expression and apoptosis during viral infection.
Collapse
Affiliation(s)
- Olivia A. Vogel
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
| | - Julianna Han
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
| | - Chieh-Yu Liang
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Santhakumar Manicassamy
- Cancer Immunology, Inflammation, and Tolerance Program, GRU Cancer Center, Augusta University, Augusta, Georgia
| | - Jasmine T. Perez
- Department of Microbiology, University of Chicago, Chicago, Illinois, United States of America
| | - Balaji Manicassamy
- Department of Microbiology and Immunology, University of Iowa, Iowa City, Iowa, United States of America
| |
Collapse
|
14
|
Li Y, Yang X, Wang N, Wang H, Yin B, Yang X, Jiang W. The divergence between SARS-CoV-2 and RaTG13 might be overestimated due to the extensive RNA modification. Future Virol 2020. [PMCID: PMC7346715 DOI: 10.2217/fvl-2020-0066] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Aim: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread throughout the world. There is urgent need to understand the phylogeny, divergence and origin of SARS-CoV-2. Materials & methods: A recent study claimed that there was 17% divergence between SARS-CoV-2 and RaTG13 (a SARS-related coronaviruses) on synonymous sites by using sequence alignment. We re-analyzed the sequences of the two coronaviruses with the same methodology. Results: We found that 87% of the synonymous substitutions between the two coronaviruses could be potentially explained by the RNA modification system in hosts, with 65% contributed by deamination on cytidines (C-T mismatches) and 22% contributed by deamination on adenosines (A-G mismatches). Conclusion: Our results demonstrate that the divergence between SARS-CoV-2 and RaTG13 has been overestimated.
Collapse
Affiliation(s)
- Yue Li
- Department of Respiratory Diseases, Qingdao Haici Hospital, PR China
| | - Xinai Yang
- Department of Respiratory Diseases, Qingdao Haici Hospital, PR China
| | - Na Wang
- Department of Respiratory Diseases, Qingdao Haici Hospital, PR China
| | - Haiyan Wang
- Department of Respiratory Diseases, Qingdao Haici Hospital, PR China
| | - Bin Yin
- Department of Respiratory Diseases, Qingdao Haici Hospital, PR China
| | - Xiaoping Yang
- Department of Respiratory Diseases, Qingdao Haici Hospital, PR China
| | - Wenqing Jiang
- Department of Respiratory Diseases, Qingdao Haici Hospital, PR China
| |
Collapse
|
15
|
Di Giorgio S, Martignano F, Torcia MG, Mattiuz G, Conticello SG. Evidence for host-dependent RNA editing in the transcriptome of SARS-CoV-2. SCIENCE ADVANCES 2020; 6:eabb5813. [PMID: 32596474 PMCID: PMC7299625 DOI: 10.1126/sciadv.abb5813] [Citation(s) in RCA: 271] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/05/2020] [Indexed: 05/13/2023]
Abstract
The COVID-19 outbreak has become a global health risk, and understanding the response of the host to the SARS-CoV-2 virus will help to combat the disease. RNA editing by host deaminases is an innate restriction process to counter virus infection, but it is not yet known whether this process operates against coronaviruses. Here, we analyze RNA sequences from bronchoalveolar lavage fluids obtained from coronavirus-infected patients. We identify nucleotide changes that may be signatures of RNA editing: adenosine-to-inosine changes from ADAR deaminases and cytosine-to-uracil changes from APOBEC deaminases. Mutational analysis of genomes from different strains of Coronaviridae from human hosts reveals mutational patterns consistent with those observed in the transcriptomic data. However, the reduced ADAR signature in these data raises the possibility that ADARs might be more effective than APOBECs in restricting viral propagation. Our results thus suggest that both APOBECs and ADARs are involved in coronavirus genome editing, a process that may shape the fate of both virus and patient.
Collapse
Affiliation(s)
- Salvatore Di Giorgio
- Core Research Laboratory, ISPRO, Firenze 50139, Italy
- Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Filippo Martignano
- Core Research Laboratory, ISPRO, Firenze 50139, Italy
- Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Maria Gabriella Torcia
- Department of Experimental and Clinical Medicine, University of Florence, Firenze 50139, Italy
| | - Giorgio Mattiuz
- Core Research Laboratory, ISPRO, Firenze 50139, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Firenze 50139, Italy
| | - Silvestro G. Conticello
- Core Research Laboratory, ISPRO, Firenze 50139, Italy
- Institute of Clinical Physiology, National Research Council, 56124 Pisa, Italy
| |
Collapse
|
16
|
Goncharov AO, Kliuchnikova AA, Nasaev SS, Moshkovskii SA. RNA Editing by ADAR Adenosine Deaminases: From Molecular Plasticity of Neural Proteins to the Mechanisms of Human Cancer. BIOCHEMISTRY (MOSCOW) 2019; 84:896-904. [PMID: 31522671 DOI: 10.1134/s0006297919080054] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
RNA editing by adenosine deaminases of the ADAR family attracts a growing interest of researchers, both zoologists studying ecological and evolutionary plasticity of invertebrates and medical biochemists focusing on the mechanisms of cancer and other human diseases. These enzymes deaminate adenosine residues in the double-stranded (ds) regions of RNA with the formation of inosine. As a result, some RNAs change their three-dimensional structure and functions. Adenosine-to-inosine editing in the mRNA coding sequences may cause amino acid substitutions in the encoded proteins. Here, we reviewed current concepts on the functions of two active ADAR isoforms identified in mammals (including humans). The ADAR1 protein, which acts non-specifically on extended dsRNA regions, is capable of immunosuppression via inactivation of the dsRNA interactions with specific sensors inducing the cell immunity. Expression of a specific ADAR1 splicing variant is regulated by the type I interferons by the negative feedback mechanism. It was shown that immunosuppressing effects of ADAR1 facilitate progression of some types of cancer. On the other hand, changes in the amino acid sequences resulting from the mRNA editing by the ADAR enzymes can result in the formation of neoantigens that can activate the antitumor immunity. The ADAR2 isoform acts on RNA more selectively; its function is associated with the editing of mRNA coding regions and can lead to the amino acid substitutions, in particular, those essential for the proper functioning of some neurotransmitter receptors in the central nervous system.
Collapse
Affiliation(s)
- A O Goncharov
- Institute of Biomedical Chemistry, Moscow, 119121, Russia.
| | - A A Kliuchnikova
- Institute of Biomedical Chemistry, Moscow, 119121, Russia.,Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - S S Nasaev
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - S A Moshkovskii
- Institute of Biomedical Chemistry, Moscow, 119121, Russia. .,Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
17
|
Christofi T, Zaravinos A. RNA editing in the forefront of epitranscriptomics and human health. J Transl Med 2019; 17:319. [PMID: 31547885 PMCID: PMC6757416 DOI: 10.1186/s12967-019-2071-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 09/17/2019] [Indexed: 12/21/2022] Open
Abstract
Post-transcriptional modifications have been recently expanded with the addition of RNA editing, which is predominantly mediated by adenosine and cytidine deaminases acting on DNA and RNA. Here, we review the full spectrum of physiological processes in which these modifiers are implicated, among different organisms. Adenosine to inosine (A-to-I) editors, members of the ADAR and ADAT protein families are important regulators of alternative splicing and transcriptional control. On the other hand, cytidine to uridine (C-to-U) editors, members of the AID/APOBEC family, are heavily implicated in innate and adaptive immunity with important roles in antibody diversification and antiviral response. Physiologically, these enzymes are present in the nucleus and/or the cytoplasm, where they modify various RNA molecules, including miRNAs, tRNAs apart from mRNAs, whereas DNA editing is also possible by some of them. The expansion of next generation sequencing technologies provided a wealth of data regarding such modifications. RNA editing has been implicated in various disorders including cancer, and neurological diseases of the brain or the central nervous system. It is also related to cancer heterogeneity and the onset of carcinogenesis. Response to treatment can also be affected by the RNA editing status where drug efficacy is significantly compromised. Studying RNA editing events can pave the way to the identification of new disease biomarkers, and provide a more personalised therapy to various diseases.
Collapse
Affiliation(s)
- Theodoulakis Christofi
- Department of Life Sciences, School of Sciences, European University Cyprus, 2404, Nicosia, Cyprus
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, 2404, Nicosia, Cyprus.
- Centre for Risk and Decision Sciences (CERIDES), 2404, Nicosia, Cyprus.
| |
Collapse
|
18
|
Dong J, Rao D, Ding Y, Zhao Y, Zhang G, Deng K, Liu T, Jiao F, Hu J, Wang H, Zhang N, Zhao P, Leng C. Hypermutations in porcine respiratory and reproductive syndrome virus. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2019; 83:104-109. [PMID: 31097872 PMCID: PMC6450159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/09/2018] [Indexed: 06/09/2023]
Abstract
Porcine reproductive and respiratory syndrome (PRRS), which is caused by the PRRS virus (PRRSV), has resulted in large economic losses for the swine industry. The virus has shown remarkable genetic diversity since its discovery. In our study, we investigated mutation types in the evolution of PRRSV for both in vivo and in vitro passaging of the virus. Sequence alignment analysis demonstrated that the most common hypermutations expressed were A→G/U→C and G→A/C→U. The data provide a new theoretical basis for PRRSV evolution.
Collapse
Affiliation(s)
- Jianguo Dong
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Dan Rao
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Yushan Ding
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Yu Zhao
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Guangqiang Zhang
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Kaiwei Deng
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Tao Liu
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Fengchao Jiao
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Jing Hu
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Huanan Wang
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Ning Zhang
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Pandeng Zhao
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| | - Chaoliang Leng
- School of Animal Husbandry and Medical Engineering, Xinyang Agriculture and Forestry University, 1 North Road, Pingqiao District, Xinyang 464000, China (Rao, Yu Zhao, Guangqiang Zhang, Deng, Liu, Jiao, Hu, Dong); Henan Key Laboratory of Insect Biology in Funiu Mountain, Henan Provincial Engineering Laboratory of Insects Bio-reactor, China-UK-NYNU-RRes Joint Laboratory of Insect Biology, Nanyang Normal University, 1638 Wolong Road, Wolong District, Nanyang 473061, China (Ding, Leng); Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China (Pandeng Zhao); Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China (Wang); Henan Fengyuan Hepu Agricultural and Animal Husbandry Co., Ltd, Zhumadian 463000, China (Ning Zhang)
| |
Collapse
|
19
|
Samuel CE. Adenosine deaminase acting on RNA (ADAR1), a suppressor of double-stranded RNA-triggered innate immune responses. J Biol Chem 2019; 294:1710-1720. [PMID: 30710018 PMCID: PMC6364763 DOI: 10.1074/jbc.tm118.004166] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Herbert "Herb" Tabor, who celebrated his 100th birthday this past year, served the Journal of Biological Chemistry as a member of the Editorial Board beginning in 1961, as an Associate Editor, and as Editor-in-Chief for 40 years, from 1971 until 2010. Among the many discoveries in biological chemistry during this period was the identification of RNA modification by C6 deamination of adenosine (A) to produce inosine (I) in double-stranded (ds) RNA. This posttranscriptional RNA modification by adenosine deamination, known as A-to-I RNA editing, diversifies the transcriptome and modulates the innate immune interferon response. A-to-I editing is catalyzed by a family of enzymes, adenosine deaminases acting on dsRNA (ADARs). The roles of A-to-I editing are varied and include effects on mRNA translation, pre-mRNA splicing, and micro-RNA silencing. Suppression of dsRNA-triggered induction and action of interferon, the cornerstone of innate immunity, has emerged as a key function of ADAR1 editing of self (cellular) and nonself (viral) dsRNAs. A-to-I modification of RNA is essential for the normal regulation of cellular processes. Dysregulation of A-to-I editing by ADAR1 can have profound consequences, ranging from effects on cell growth and development to autoimmune disorders.
Collapse
Affiliation(s)
- Charles E Samuel
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California 93106.
| |
Collapse
|
20
|
Long-read sequencing uncovers a complex transcriptome topology in varicella zoster virus. BMC Genomics 2018; 19:873. [PMID: 30514211 DOI: 10.1186/s12864-018-5267-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/19/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Varicella zoster virus (VZV) is a human pathogenic alphaherpesvirus harboring a relatively large DNA molecule. The VZV transcriptome has already been analyzed by microarray and short-read sequencing analyses. However, both approaches have substantial limitations when used for structural characterization of transcript isoforms, even if supplemented with primer extension or other techniques. Among others, they are inefficient in distinguishing between embedded RNA molecules, transcript isoforms, including splice and length variants, as well as between alternative polycistronic transcripts. It has been demonstrated in several studies that long-read sequencing is able to circumvent these problems. RESULTS In this work, we report the analysis of the VZV lytic transcriptome using the Oxford Nanopore Technologies sequencing platform. These investigations have led to the identification of 114 novel transcripts, including mRNAs, non-coding RNAs, polycistronic RNAs and complex transcripts, as well as 10 novel spliced transcripts and 25 novel transcription start site isoforms and transcription end site isoforms. A novel class of transcripts, the nroRNAs are described in this study. These transcripts are encoded by the genomic region located in close vicinity to the viral replication origin. We also show that the ORF63 exhibits a complex structural variation encompassing the splice sites of VZV latency transcripts. Additionally, we have detected RNA editing in a novel non-coding RNA molecule. CONCLUSIONS Our investigations disclosed a composite transcriptomic architecture of VZV, including the discovery of novel RNA molecules and transcript isoforms, as well as a complex meshwork of transcriptional read-throughs and overlaps. The results represent a substantial advance in the annotation of the VZV transcriptome and in understanding the molecular biology of the herpesviruses in general.
Collapse
|
21
|
Whitmer SLM, Ladner JT, Wiley MR, Patel K, Dudas G, Rambaut A, Sahr F, Prieto K, Shepard SS, Carmody E, Knust B, Naidoo D, Deen G, Formenty P, Nichol ST, Palacios G, Ströher U. Active Ebola Virus Replication and Heterogeneous Evolutionary Rates in EVD Survivors. Cell Rep 2018; 22:1159-1168. [PMID: 29386105 PMCID: PMC5809616 DOI: 10.1016/j.celrep.2018.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/20/2017] [Accepted: 01/02/2018] [Indexed: 11/18/2022] Open
Abstract
Following cessation of continuous Ebola virus (EBOV) transmission within Western Africa, sporadic EBOV disease (EVD) cases continued to re-emerge beyond the viral incubation period. Epidemiological and genomic evidence strongly suggests that this represented transmission from EVD survivors. To investigate whether persistent infections are characterized by ongoing viral replication, we sequenced EBOV from the semen of nine EVD survivors and a subset of corresponding acute specimens. EBOV evolutionary rates during persistence were either similar to or reduced relative to acute infection rates. Active EBOV replication/transcription continued during convalescence, but decreased over time, consistent with viral persistence rather than viral latency. Patterns of genetic divergence suggest a moderate relaxation of selective constraints within the sGP carboxy-terminal tail during persistent infections, but do not support widespread diversifying selection. Altogether, our data illustrate that EBOV persistence in semen, urine, and aqueous humor is not a quiescent or latent infection.
Collapse
Affiliation(s)
- Shannon L M Whitmer
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Jason T Ladner
- Center for Genome Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Michael R Wiley
- Center for Genome Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Ketan Patel
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Gytis Dudas
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Andrew Rambaut
- Institute of Evolutionary Biology, University of Edinburgh, King's Buildings, Edinburgh, UK; Fogarty International Center, National Institutes of Health, Bethesda, MD, USA
| | - Foday Sahr
- Sierra Leone Armed Forces, Freetown, Sierra Leone
| | - Karla Prieto
- Center for Genome Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Samuel S Shepard
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Ellie Carmody
- Division of Infectious Diseases, NYU School of Medicine, Bellevue Hospital Center, New York, NY, USA
| | - Barbara Knust
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Dhamari Naidoo
- Health Emergency Programme, World Health Organization, Geneva, Switzerland
| | - Gibrilla Deen
- Sierra Leone Ministry of Health and Sanitation, Freetown, Sierra Leone
| | - Pierre Formenty
- Health Emergency Programme, World Health Organization, Geneva, Switzerland
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Gustavo Palacios
- Center for Genome Sciences, US Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Ute Ströher
- Viral Special Pathogens Branch, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
22
|
Epitranscriptomic regulation of viral replication. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:460-471. [PMID: 28219769 DOI: 10.1016/j.bbagrm.2017.02.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/10/2017] [Accepted: 02/10/2017] [Indexed: 12/11/2022]
Abstract
RNA plays central roles in biology and novel functions and regulation mechanisms are constantly emerging. To accomplish some of their functions within the cell, RNA molecules undergo hundreds of chemical modifications from which N6-methyladenosine (m6A), inosine (I), pseudouridine (ψ) and 5-methylcytosine (5mC) have been described in eukaryotic mRNA. Interestingly, the m6A modification was shown to be reversible, adding novel layers of regulation of gene expression through what is now recognized as epitranscriptomics. The development of molecular mapping strategies coupled to next generation sequencing allowed the identification of thousand of modified transcripts in different tissues and under different physiological conditions such as viral infections. As intracellular parasites, viruses are confronted to cellular RNA modifying enzymes and, as a consequence, viral RNA can be chemically modified at some stages of the replication cycle. This review focuses on the chemical modifications of viral RNA and the impact that these modifications have on viral gene expression and the output of infection. A special emphasis is given to m6A, which was recently shown to play important yet controversial roles in different steps of the HIV-1, HCV and ZIKV replication cycles.
Collapse
|
23
|
Sanjuán R, Domingo-Calap P. Mechanisms of viral mutation. Cell Mol Life Sci 2016; 73:4433-4448. [PMID: 27392606 PMCID: PMC5075021 DOI: 10.1007/s00018-016-2299-6] [Citation(s) in RCA: 526] [Impact Index Per Article: 58.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/20/2016] [Accepted: 06/22/2016] [Indexed: 02/08/2023]
Abstract
The remarkable capacity of some viruses to adapt to new hosts and environments is highly dependent on their ability to generate de novo diversity in a short period of time. Rates of spontaneous mutation vary amply among viruses. RNA viruses mutate faster than DNA viruses, single-stranded viruses mutate faster than double-strand virus, and genome size appears to correlate negatively with mutation rate. Viral mutation rates are modulated at different levels, including polymerase fidelity, sequence context, template secondary structure, cellular microenvironment, replication mechanisms, proofreading, and access to post-replicative repair. Additionally, massive numbers of mutations can be introduced by some virus-encoded diversity-generating elements, as well as by host-encoded cytidine/adenine deaminases. Our current knowledge of viral mutation rates indicates that viral genetic diversity is determined by multiple virus- and host-dependent processes, and that viral mutation rates can evolve in response to specific selective pressures.
Collapse
Affiliation(s)
- Rafael Sanjuán
- Department of Genetics and Institute for Integrative Systems Biology (I2SysBio), Universitat de València, C/Catedrático José Beltrán 2, 46980, Paterna, Valencia, Spain.
| | - Pilar Domingo-Calap
- Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, LabEx Transplantex, Centre de Recherche d'Immunologie et d'Hématologie, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France
- Fédération Hospitalo-Universitaire OMICARE, Centre de Recherche d'Immunologie et d'Hématologie, Strasbourg, France
| |
Collapse
|
24
|
Orecchini E, Doria M, Antonioni A, Galardi S, Ciafrè SA, Frassinelli L, Mancone C, Montaldo C, Tripodi M, Michienzi A. ADAR1 restricts LINE-1 retrotransposition. Nucleic Acids Res 2016; 45:155-168. [PMID: 27658966 PMCID: PMC5224506 DOI: 10.1093/nar/gkw834] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/08/2016] [Accepted: 09/09/2016] [Indexed: 12/16/2022] Open
Abstract
Adenosine deaminases acting on RNA (ADARs) are involved in RNA editing that converts adenosines to inosines in double-stranded RNAs. ADAR1 was demonstrated to be functional on different viruses exerting either antiviral or proviral effects. Concerning HIV-1, several studies showed that ADAR1 favors viral replication. The aim of this study was to investigate the composition of the ADAR1 ribonucleoprotein complex during HIV-1 expression. By using a dual-tag affinity purification procedure in cells expressing HIV-1 followed by mass spectrometry analysis, we identified 14 non-ribosomal ADAR1-interacting proteins, most of which are novel. A significant fraction of these proteins were previously demonstrated to be associated to the Long INterspersed Element 1 (LINE1 or L1) ribonucleoparticles and to regulate the life cycle of L1 retrotransposons that continuously re-enter host-genome. Hence, we investigated the function of ADAR1 in the regulation of L1 activity. By using different cell-culture based retrotransposition assays in HeLa cells, we demonstrated a novel function of ADAR1 as suppressor of L1 retrotransposition. Apparently, this inhibitory mechanism does not occur through ADAR1 editing activity. Furthermore, we showed that ADAR1 binds the basal L1 RNP complex. Overall, these data support the role of ADAR1 as regulator of L1 life cycle.
Collapse
Affiliation(s)
- Elisa Orecchini
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, Rome 00133, Italy
| | - Margherita Doria
- Laboratory of Immunoinfectivology, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, Rome 00165, Italy
| | - Ambra Antonioni
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, Rome 00133, Italy
| | - Silvia Galardi
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, Rome 00133, Italy
| | - Silvia Anna Ciafrè
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, Rome 00133, Italy
| | - Loredana Frassinelli
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, Rome 00133, Italy
| | - Carmine Mancone
- Istituto Pasteur-Fondazione Cenci Bolognetti, Department of Cellular Biotechnologies and Haematology, Sapienza University of Rome, Italy.,L. Spallanzani National Institute for Infectious Diseases, IRCCS, via Portuense 292, Rome 00149, Italy
| | - Claudia Montaldo
- L. Spallanzani National Institute for Infectious Diseases, IRCCS, via Portuense 292, Rome 00149, Italy
| | - Marco Tripodi
- Istituto Pasteur-Fondazione Cenci Bolognetti, Department of Cellular Biotechnologies and Haematology, Sapienza University of Rome, Italy.,L. Spallanzani National Institute for Infectious Diseases, IRCCS, via Portuense 292, Rome 00149, Italy
| | - Alessandro Michienzi
- Department of Biomedicine and Prevention, University of Rome 'Tor Vergata', Via Montpellier 1, Rome 00133, Italy
| |
Collapse
|
25
|
Figueroa T, Boumart I, Coupeau D, Rasschaert D. Hyperediting by ADAR1 of a new herpesvirus lncRNA during the lytic phase of the oncogenic Marek's disease virus. J Gen Virol 2016; 97:2973-2988. [PMID: 27655063 DOI: 10.1099/jgv.0.000606] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Marek's disease virus, or Gallid herpesvirus 2 (GaHV-2), is an avian alphaherpesvirus that induces T-cell lymphoma in chickens. During transcriptomic studies of the RL region of the genome, we characterized the 7.5 kbp gene of the ERL lncRNA (edited repeat-long, long non-coding RNA), which may act as a natural antisense transcript (NAT) of the major GaHV-2 oncogene meq and of two of the three miRNA clusters. During infections in vivo and in vitro, we detected hyperediting of the ERL lncRNA that appeared to be directly correlated with ADAR1 expression levels. The ERL lncRNA was expressed equally during the lytic and latent phases of infection and during viral reactivation, but its hyperediting increased only during the lytic infection of chicken embryo fibroblasts. We also showed that chicken ADAR1 expression was controlled by the JAK/STAT IFN-response pathway, through an inducible promoter containing IFN-stimulated response elements that were functional during stimulation with IFN-α or poly(I:C). Like the human and murine miR-155-5p, the chicken gga-miR-155-5p and the GaHV-2 analogue mdv1-miR-M4-5p deregulated this pathway by targeting and repressing expression of suppressor of cytokine signalling 1, leading to the upregulation of ADAR1. Finally, we hypothesized that the natural antisense transcript role of the ERL lncRNA could be disrupted by its hyperediting, particularly during viral lytic replication, and that the observed deregulation of the innate immune system by mdv1-miR-M4-5p might contribute to the viral cycle.
Collapse
Affiliation(s)
- Thomas Figueroa
- Equipe Transcription et Lymphome Viro-Induit (TLVI), UMR 7261 CNRS/Université François Rabelais de Tours, Tours, France
| | - Imane Boumart
- Equipe Transcription et Lymphome Viro-Induit (TLVI), UMR 7261 CNRS/Université François Rabelais de Tours, Tours, France
| | - Damien Coupeau
- Equipe Transcription et Lymphome Viro-Induit (TLVI), UMR 7261 CNRS/Université François Rabelais de Tours, Tours, France
| | - Denis Rasschaert
- Equipe Transcription et Lymphome Viro-Induit (TLVI), UMR 7261 CNRS/Université François Rabelais de Tours, Tours, France
| |
Collapse
|
26
|
Cuevas JM, Combe M, Torres-Puente M, Garijo R, Guix S, Buesa J, Rodríguez-Díaz J, Sanjuán R. Human norovirus hyper-mutation revealed by ultra-deep sequencing. INFECTION GENETICS AND EVOLUTION 2016; 41:233-239. [PMID: 27094861 PMCID: PMC7172324 DOI: 10.1016/j.meegid.2016.04.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/11/2016] [Accepted: 04/15/2016] [Indexed: 02/02/2023]
Abstract
Human noroviruses (NoVs) are a major cause of gastroenteritis worldwide. It is thought that, similar to other RNA viruses, high mutation rates allow NoVs to evolve fast and to undergo rapid immune escape at the population level. However, the rate and spectrum of spontaneous mutations of human NoVs have not been quantified previously. Here, we analyzed the intra-patient diversity of the NoV capsid by carrying out RT-PCR and ultra-deep sequencing with 100,000-fold coverage of 16 stool samples from symptomatic patients. This revealed the presence of low-frequency sequences carrying large numbers of U-to-C or A-to-G base transitions, suggesting a role for hyper-mutation in NoV diversity. To more directly test for hyper-mutation, we performed transfection assays in which the production of mutations was restricted to a single cell infection cycle. This confirmed the presence of sequences with multiple U-to-C/A-to-G transitions, and suggested that hyper-mutation contributed a large fraction of the total NoV spontaneous mutation rate. The type of changes produced and their sequence context are compatible with ADAR-mediated editing of the viral RNA. Norovirus U-to-C hyper-mutants are present in patient samples. Analysis of hyper-mutants in cell culture suggests ADAR-mediated RNA edition. Hyper-mutation may contribute to norovirus diversity and evolution.
Collapse
Affiliation(s)
- José M Cuevas
- Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Universitat de València, Valencia, Spain
| | - Marine Combe
- Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Universitat de València, Valencia, Spain
| | - Manoli Torres-Puente
- Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana, Valencia, Spain
| | - Raquel Garijo
- Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Universitat de València, Valencia, Spain
| | - Susana Guix
- Departament de Microbiologia, Universitat de Barcelona, Barcelona, Spain
| | - Javier Buesa
- Departament de Microbiologia, Universitat de València, Valencia, Spain
| | | | - Rafael Sanjuán
- Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Universitat de València, Valencia, Spain; Departament de Genètica, Universitat de València, Valencia, Spain.
| |
Collapse
|
27
|
Abstract
The family Arenaviridae currently comprises over 20 viral species, each of them associated with a main rodent species as the natural reservoir and in one case possibly phyllostomid bats. Moreover, recent findings have documented a divergent group of arenaviruses in captive alethinophidian snakes. Human infections occur through mucosal exposure to aerosols or by direct contact of abraded skin with infectious materials. Arenaviruses merit interest both as highly tractable experimental model systems to study acute and persistent infections and as clinically important human pathogens including Lassa (LASV) and Junin (JUNV) viruses, the causative agents of Lassa and Argentine hemorrhagic fevers (AHFs), respectively, for which there are no FDA-licensed vaccines, and current therapy is limited to an off-label use of ribavirin (Rib) that has significant limitations. Arenaviruses are enveloped viruses with a bi-segmented negative strand (NS) RNA genome. Each genome segment, L (ca 7.3 kb) and S (ca 3.5 kb), uses an ambisense coding strategy to direct the synthesis of two polypeptides in opposite orientation, separated by a noncoding intergenic region (IGR). The S genomic RNA encodes the virus nucleoprotein (NP) and the precursor (GPC) of the virus surface glycoprotein that mediates virus receptor recognition and cell entry via endocytosis. The L genome RNA encodes the viral RNA-dependent RNA polymerase (RdRp, or L polymerase) and the small (ca 11 kDa) RING finger protein Z that has functions of a bona fide matrix protein including directing virus budding. Arenaviruses were thought to be relatively stable genetically with intra- and interspecies amino acid sequence identities of 90-95 % and 44-63 %, respectively. However, recent evidence has documented extensive arenavirus genetic variability in the field. Moreover, dramatic phenotypic differences have been documented among closely related LCMV isolates. These data provide strong evidence of viral quasispecies involvement in arenavirus adaptability and pathogenesis. Here, we will review several aspects of the molecular biology of arenaviruses, phylogeny and evolution, and quasispecies dynamics of arenavirus populations for a better understanding of arenavirus pathogenesis, as well as for the development of novel antiviral strategies to combat arenavirus infections.
Collapse
Affiliation(s)
- Esteban Domingo
- Campus de Cantoblanco, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
| | - Peter Schuster
- The Santa Fe Institute, Santa Fe, NM, USA and Institut f. Theoretische Chemie, Universität Wien, Vienna, Austria
| |
Collapse
|
28
|
Deffit SN, Hundley HA. To edit or not to edit: regulation of ADAR editing specificity and efficiency. WILEY INTERDISCIPLINARY REVIEWS-RNA 2015; 7:113-27. [PMID: 26612708 DOI: 10.1002/wrna.1319] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/15/2015] [Accepted: 10/20/2015] [Indexed: 11/08/2022]
Abstract
Hundreds to millions of adenosine (A)-to-inosine (I) modifications are present in eukaryotic transcriptomes and play an essential role in the creation of proteomic and phenotypic diversity. As adenosine and inosine have different base-pairing properties, the functional consequences of these modifications or 'edits' include altering coding potential, splicing, and miRNA-mediated gene silencing of transcripts. However, rather than serving as a static control of gene expression, A-to-I editing provides a means to dynamically rewire the genetic code during development and in a cell-type specific manner. Interestingly, during normal development, in specific cells, and in both neuropathological diseases and cancers, the extent of RNA editing does not directly correlate with levels of the substrate mRNA or the adenosine deaminase that act on RNA (ADAR) editing enzymes, implying that cellular factors are required for spatiotemporal regulation of A-to-I editing. The factors that affect the specificity and extent of ADAR activity have been thoroughly dissected in vitro. Yet, we still lack a complete understanding of how specific ADAR family members can selectively deaminate certain adenosines while others cannot. Additionally, in the cellular environment, ADAR specificity and editing efficiency is likely to be influenced by cellular factors, which is currently an area of intense investigation. Data from many groups have suggested two main mechanisms for controlling A-to-I editing in the cell: (1) regulating ADAR accessibility to target RNAs and (2) protein-protein interactions that directly alter ADAR enzymatic activity. Recent studies suggest cis- and trans-acting RNA elements, heterodimerization and RNA-binding proteins play important roles in regulating RNA editing levels in vivo. WIREs RNA 2016, 7:113-127. doi: 10.1002/wrna.1319.
Collapse
Affiliation(s)
- Sarah N Deffit
- Medical Sciences Program, Indiana University, Bloomington, IN, USA
| | | |
Collapse
|
29
|
Cuevas JM, Geller R, Garijo R, López-Aldeguer J, Sanjuán R. Extremely High Mutation Rate of HIV-1 In Vivo. PLoS Biol 2015; 13:e1002251. [PMID: 26375597 PMCID: PMC4574155 DOI: 10.1371/journal.pbio.1002251] [Citation(s) in RCA: 251] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 08/10/2015] [Indexed: 11/18/2022] Open
Abstract
Rates of spontaneous mutation critically determine the genetic diversity and evolution of RNA viruses. Although these rates have been characterized in vitro and in cell culture models, they have seldom been determined in vivo for human viruses. Here, we use the intrapatient frequency of premature stop codons to quantify the HIV-1 genome-wide rate of spontaneous mutation in DNA sequences from peripheral blood mononuclear cells. This reveals an extremely high mutation rate of (4.1 ± 1.7) × 10−3 per base per cell, the highest reported for any biological entity. Sequencing of plasma-derived sequences yielded a mutation frequency 44 times lower, indicating that a large fraction of viral genomes are lethally mutated and fail to reach plasma. We show that the HIV-1 reverse transcriptase contributes only 2% of mutations, whereas 98% result from editing by host cytidine deaminases of the A3 family. Hypermutated viral sequences are less abundant in patients showing rapid disease progression compared to normal progressors, highlighting the antiviral role of A3 proteins. However, the amount of A3-mediated editing varies broadly, and we find that low-edited sequences are more abundant among rapid progressors, suggesting that suboptimal A3 activity might enhance HIV-1 genetic diversity and pathogenesis. The rate of spontaneous mutation of the HIV-1 genome within its human host is exceptionally high, is mostly driven by host cytidine deaminases, and probably plays a role in disease progression. The high levels of genetic diversity of the HIV-1 virus grant it the ability to escape the immune system, to rapidly evolve drug resistance, and to circumvent vaccination strategies. However, our knowledge of HIV-1 mutation rates has been largely restricted to in vitro and cell culture studies because of the inherent complexity of measuring these rates in vivo. Here, by analyzing the frequency of premature stop codons, we show that the HIV-1 mutation rate in vivo is two orders of magnitude higher than that predicted by in vitro studies, making it the highest reported mutation rate for any biological system. A large component of this rate is from host cellular cytidine deaminases, which induce mutations in the viral DNA as a defense mechanism. While the HIV-1 genome is hypermutated in blood cells, only a very small fraction of these mutations reach the plasma, indicating that many viruses are defective as a result of the extremely high mutation load. In addition, we find that the HIV-1 mutation rate tends to be higher in patients showing normal disease progression than in those undergoing rapid progression, emphasizing the negative impact on viral fitness of hypermutation by host cytidine deaminases. However, we also observe subpopulations of weakly-mutated viral genomes whose sequence diversity may influence viral pathogenesis. Our work highlights the fine balance for HIV-1 between enough mutation to evade host responses and too much mutation that can inactivate the virus.
Collapse
Affiliation(s)
- José M. Cuevas
- Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Valencia, Spain
| | - Ron Geller
- Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Valencia, Spain
| | - Raquel Garijo
- Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Valencia, Spain
| | - José López-Aldeguer
- Hospital Universitario La Fe, Valencia, Spain
- CoRIS and HIV Biobank, Spanish AIDS Research Network, Spain
| | - Rafael Sanjuán
- Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Valencia, Spain
- Departament de Genètica, Universitat de València, Valencia, Spain
- * E-mail:
| |
Collapse
|
30
|
Park DJ, Dudas G, Wohl S, Goba A, Whitmer SLM, Andersen KG, Sealfon RS, Ladner JT, Kugelman JR, Matranga CB, Winnicki SM, Qu J, Gire SK, Gladden-Young A, Jalloh S, Nosamiefan D, Yozwiak NL, Moses LM, Jiang PP, Lin AE, Schaffner SF, Bird B, Towner J, Mamoh M, Gbakie M, Kanneh L, Kargbo D, Massally JLB, Kamara FK, Konuwa E, Sellu J, Jalloh AA, Mustapha I, Foday M, Yillah M, Erickson BR, Sealy T, Blau D, Paddock C, Brault A, Amman B, Basile J, Bearden S, Belser J, Bergeron E, Campbell S, Chakrabarti A, Dodd K, Flint M, Gibbons A, Goodman C, Klena J, McMullan L, Morgan L, Russell B, Salzer J, Sanchez A, Wang D, Jungreis I, Tomkins-Tinch C, Kislyuk A, Lin MF, Chapman S, MacInnis B, Matthews A, Bochicchio J, Hensley LE, Kuhn JH, Nusbaum C, Schieffelin JS, Birren BW, Forget M, Nichol ST, Palacios GF, Ndiaye D, Happi C, Gevao SM, Vandi MA, Kargbo B, Holmes EC, Bedford T, Gnirke A, Ströher U, Rambaut A, Garry RF, Sabeti PC. Ebola Virus Epidemiology, Transmission, and Evolution during Seven Months in Sierra Leone. Cell 2015; 161:1516-26. [PMID: 26091036 PMCID: PMC4503805 DOI: 10.1016/j.cell.2015.06.007] [Citation(s) in RCA: 224] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 05/26/2015] [Accepted: 06/01/2015] [Indexed: 11/29/2022]
Abstract
The 2013-2015 Ebola virus disease (EVD) epidemic is caused by the Makona variant of Ebola virus (EBOV). Early in the epidemic, genome sequencing provided insights into virus evolution and transmission and offered important information for outbreak response. Here, we analyze sequences from 232 patients sampled over 7 months in Sierra Leone, along with 86 previously released genomes from earlier in the epidemic. We confirm sustained human-to-human transmission within Sierra Leone and find no evidence for import or export of EBOV across national borders after its initial introduction. Using high-depth replicate sequencing, we observe both host-to-host transmission and recurrent emergence of intrahost genetic variants. We trace the increasing impact of purifying selection in suppressing the accumulation of nonsynonymous mutations over time. Finally, we note changes in the mucin-like domain of EBOV glycoprotein that merit further investigation. These findings clarify the movement of EBOV within the region and describe viral evolution during prolonged human-to-human transmission.
Collapse
Affiliation(s)
- Daniel J Park
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA.
| | - Gytis Dudas
- Institute of Evolutionary Biology, Ashworth Laboratories, University of Edinburgh, Edinburgh EH9 3FL, UK
| | - Shirlee Wohl
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA; Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | | | - Shannon L M Whitmer
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Kristian G Andersen
- Scripps Translational Science Institute, The Scripps Research Institute, 3344 N Torrey Pines Court, La Jolla, CA 92037, USA
| | - Rachel S Sealfon
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA; Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Jason T Ladner
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Jeffrey R Kugelman
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | | | - Sarah M Winnicki
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA; Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - James Qu
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA
| | - Stephen K Gire
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA; Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | | | | | - Dolo Nosamiefan
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA
| | - Nathan L Yozwiak
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA; Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - Lina M Moses
- Tulane University, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Pan-Pan Jiang
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA; Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - Aaron E Lin
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA; Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - Stephen F Schaffner
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA; Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - Brian Bird
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Jonathan Towner
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Mambu Mamoh
- Kenema Government Hospital, Kenema, Sierra Leone
| | | | | | - David Kargbo
- Kenema Government Hospital, Kenema, Sierra Leone
| | | | | | - Edwin Konuwa
- Kenema Government Hospital, Kenema, Sierra Leone
| | | | | | | | - Momoh Foday
- Kenema Government Hospital, Kenema, Sierra Leone
| | | | - Bobbie R Erickson
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Tara Sealy
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Dianna Blau
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Christopher Paddock
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Aaron Brault
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Brian Amman
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Jane Basile
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Scott Bearden
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Jessica Belser
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Eric Bergeron
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Shelley Campbell
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Ayan Chakrabarti
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Kimberly Dodd
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Mike Flint
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Aridth Gibbons
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Christin Goodman
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - John Klena
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Laura McMullan
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Laura Morgan
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Brandy Russell
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Johanna Salzer
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Angela Sanchez
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - David Wang
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Irwin Jungreis
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | | | - Andrey Kislyuk
- DNAnexus, 1975 West El Camino Real, Suite 101, Mountain View, CA 94040, USA
| | - Michael F Lin
- DNAnexus, 1975 West El Camino Real, Suite 101, Mountain View, CA 94040, USA
| | - Sinead Chapman
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA
| | - Bronwyn MacInnis
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA
| | - Ashley Matthews
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA; Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - James Bochicchio
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA
| | - Lisa E Hensley
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, B-8200 Research Plaza, Fort Detrick, Frederick, MD 21702, USA
| | - Chad Nusbaum
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA
| | - John S Schieffelin
- Tulane University, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Bruce W Birren
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA
| | - Marc Forget
- Médecins Sans Frontières, Rue de l'Arbre Bénit 46, 1050 Bruxelles, Belgium
| | - Stuart T Nichol
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Gustavo F Palacios
- US Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, Frederick, MD 21702, USA
| | - Daouda Ndiaye
- Université Cheikh Anta Diop, BP 5005, Dakar, Sénégal
| | - Christian Happi
- Redeemers University Nigeria, KM 46 Lagos-Ibadan Expressway, Redemption City, Ogun State, Nigeria
| | - Sahr M Gevao
- University of Sierra Leone, A.J. Momoh St, Tower Hill, Freetown, Sierra Leone
| | - Mohamed A Vandi
- Sierra Leone Ministry of Health and Sanitation, Youyi Building, Freetown, Sierra Leone
| | - Brima Kargbo
- Sierra Leone Ministry of Health and Sanitation, Youyi Building, Freetown, Sierra Leone
| | - Edward C Holmes
- University of Sydney, Johns Hopkins Drive, Camperdown NSW 2050, Australia
| | - Trevor Bedford
- Fred Hutchinson Cancer Research Center, 110 Fairview Avenue North, Seattle, WA 98109, USA
| | - Andreas Gnirke
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA
| | - Ute Ströher
- National Center for Emerging and Zoonotic Infectious Diseases and National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road NE, Mailstop-G14, Atlanta, GA 30333, USA
| | - Andrew Rambaut
- Institute of Evolutionary Biology, Ashworth Laboratories, University of Edinburgh, Edinburgh EH9 3FL, UK; Centre for Immunology, Infection and Evolution, University of Edinburgh, Ashworth Laboratories, Edinburgh EH9 3FL, UK; Fogarty International Center, National Institutes of Health, 31 Center Drive, MSC 2220 Bethesda, MD 20892, USA.
| | - Robert F Garry
- Tulane University, 1430 Tulane Avenue, SL-38, New Orleans, LA 70112, USA
| | - Pardis C Sabeti
- Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA 02142, USA; Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA.
| |
Collapse
|
31
|
Li MMH, MacDonald MR, Rice CM. To translate, or not to translate: viral and host mRNA regulation by interferon-stimulated genes. Trends Cell Biol 2015; 25:320-9. [PMID: 25748385 PMCID: PMC4441850 DOI: 10.1016/j.tcb.2015.02.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 01/28/2015] [Accepted: 02/05/2015] [Indexed: 01/08/2023]
Abstract
Translational regulation of viral and host mRNA is an important function of a subset of interferon-stimulated genes (ISGs). These ISGs repress translation through binding to viral RNA, direct interaction with, or perturbation of the translation machinery components. Several ISGs localize to cytoplasmic granules such as stress granules (SGs) and processing bodies (PBs), and interfere with the processing or function of microRNAs (miRNAs).
Type I interferon (IFN) is one of the first lines of cellular defense against viral pathogens. As a result of IFN signaling, a wide array of IFN-stimulated gene (ISG) products is upregulated to target different stages of the viral life cycle. We review recent findings implicating a subset of ISGs in translational regulation of viral and host mRNAs. Translation inhibition is mediated either by binding to viral RNA or by disrupting physiological interactions or levels of the translation complex components. In addition, many of these ISGs localize to translationally silent cytoplasmic granules, such as stress granules and processing bodies, and intersect with the microRNA (miRNA)-mediated silencing pathway to regulate translation of cellular mRNAs.
Collapse
Affiliation(s)
- Melody M H Li
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA.
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| |
Collapse
|
32
|
Double-stranded RNA-specific adenosine deaminase 1 (ADAR1) promotes EIAV replication and infectivity. Virology 2015; 476:364-371. [PMID: 25589239 DOI: 10.1016/j.virol.2014.12.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 10/23/2014] [Accepted: 12/23/2014] [Indexed: 12/23/2022]
Abstract
Adenosine deaminases that act on RNA (ADARs) have been reported to be functional on various viruses. ADAR1 may exhibit antiviral or proviral activity depending on the type of virus. Human immunodeficiency virus (HIV)-1 is the most well-studied lentivirus with respect to its interaction with ADAR1, and variable results have been reported. In this study, we demonstrated that equine ADAR1 (eADAR1) was a positive regulator of equine infectious anemia virus (EIAV), another lentivirus of the Retroviridae family. First, eADAR1 significantly promoted EIAV replication, and the enhancement of viral protein expression was associated with the long terminal repeat (LTR) and Rev response element (RRE) regions. Second, the RNA binding domain 1 of eADAR1 was essential only for enhancing LTR-mediated gene expression. Third, in contrast with APOBEC proteins, which have been shown to reduce lentiviral infectivity, eADAR1 increased the EIAV infectivity. This study indicated that eADAR1 was proviral rather than antiviral for EIAV.
Collapse
|
33
|
Cheung PPH, Rogozin IB, Choy KT, Ng HY, Peiris JSM, Yen HL. Comparative mutational analyses of influenza A viruses. RNA (NEW YORK, N.Y.) 2015; 21:36-47. [PMID: 25404565 PMCID: PMC4274636 DOI: 10.1261/rna.045369.114] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The error-prone RNA-dependent RNA polymerase (RdRP) and external selective pressures are the driving forces for RNA viral diversity. When confounded by selective pressures, it is difficult to assess if influenza A viruses (IAV) that have a wide host range possess comparable or distinct spontaneous mutational frequency in their RdRPs. We used in-depth bioinformatics analyses to assess the spontaneous mutational frequencies of two RdRPs derived from human seasonal (A/Wuhan/359/95; Wuhan) and H5N1 (A/Vietnam/1203/04; VN1203) viruses using the mini-genome system with a common firefly luciferase reporter serving as the template. High-fidelity reverse transcriptase was applied to generate high-quality mutational spectra which allowed us to assess and compare the mutational frequencies and mutable motifs along a target sequence of the two RdRPs of two different subtypes. We observed correlated mutational spectra (τ correlation P < 0.0001), comparable mutational frequencies (H3N2:5.8 ± 0.9; H5N1:6.0 ± 0.5), and discovered a highly mutable motif "(A)AAG" for both Wuhan and VN1203 RdRPs. Results were then confirmed with two recombinant A/Puerto Rico/8/34 (PR8) viruses that possess RdRP derived from Wuhan or VN1203 (RG-PR8×Wuhan(PB2, PB1, PA, NP) and RG-PR8×VN1203(PB2, PB1, PA, NP)). Applying novel bioinformatics analysis on influenza mutational spectra, we provide a platform for a comprehensive analysis of the spontaneous mutation spectra for an RNA virus.
Collapse
Affiliation(s)
- Peter Pak-Hang Cheung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Igor B Rogozin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland 20894-6075, USA
| | - Ka-Tim Choy
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hoi Yee Ng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Joseph Sriyal Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hui-Ling Yen
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
34
|
Liu Y, Ma T, Liu J, Zhao X, Cheng Z, Guo H, Xu R, Wang S. Circulating type 1 vaccine-derived poliovirus may evolve under the pressure of adenosine deaminases acting on RNA. J Matern Fetal Neonatal Med 2014; 28:2096-2099. [PMID: 25330844 DOI: 10.3109/14767058.2014.979147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 10/17/2014] [Indexed: 11/13/2022]
Abstract
Poliovirus, the causative agent of poliomyelitis, is a human enterovirus and member of the Picornaviridae family. An effective live-attenuated poliovirus vaccine strain (Sabin 1) has been developed and has protected humans from polio. However, a few cases of vaccine virulence reversion have been documented in several countries. For instance, circulating type 1 vaccine-derived poliovirus is a highly pathogenic poliovirus that evolved from an avirulent strain, but the mechanism by which vaccine strains undergo reversion remains unclear. In this study, vaccine strains exhibited A to G/U to C and G to A/C to U hypermutations in the reversed evolution of Sabin 1. Furthermore, the mutation ratios of U to C and C to U were higher than those of other mutation types. Dinucleotide editing context was then analyzed. Results showed that A to G and U to C mutations exhibited preferences similar to adenosine deaminases acting on RNA (ADAR). Hence, ADARs may participate in poliovirus vaccine evolution.
Collapse
Affiliation(s)
- Yanhan Liu
- a College of Veterinary Medicine, Research Center for Animal Disease Control Engineering Shandong Province , Shandong Agricultural University , Tai`an , China
| | - Tengfei Ma
- a College of Veterinary Medicine, Research Center for Animal Disease Control Engineering Shandong Province , Shandong Agricultural University , Tai`an , China
| | - Jianzhu Liu
- a College of Veterinary Medicine, Research Center for Animal Disease Control Engineering Shandong Province , Shandong Agricultural University , Tai`an , China
| | - Xiaona Zhao
- a College of Veterinary Medicine, Research Center for Animal Disease Control Engineering Shandong Province , Shandong Agricultural University , Tai`an , China
| | - Ziqiang Cheng
- a College of Veterinary Medicine, Research Center for Animal Disease Control Engineering Shandong Province , Shandong Agricultural University , Tai`an , China
| | - Huijun Guo
- a College of Veterinary Medicine, Research Center for Animal Disease Control Engineering Shandong Province , Shandong Agricultural University , Tai`an , China
| | - Ruixue Xu
- a College of Veterinary Medicine, Research Center for Animal Disease Control Engineering Shandong Province , Shandong Agricultural University , Tai`an , China
| | - Shujing Wang
- a College of Veterinary Medicine, Research Center for Animal Disease Control Engineering Shandong Province , Shandong Agricultural University , Tai`an , China
| |
Collapse
|
35
|
Cachat A, Alais S, Chevalier SA, Journo C, Fusil F, Dutartre H, Boniface A, Ko NL, Gessain A, Cosset FL, Suspène R, Vartanian JP, Mahieux R. ADAR1 enhances HTLV-1 and HTLV-2 replication through inhibition of PKR activity. Retrovirology 2014; 11:93. [PMID: 25389016 PMCID: PMC4245799 DOI: 10.1186/s12977-014-0093-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 10/16/2014] [Indexed: 11/25/2022] Open
Abstract
Background The role of innate immunity in general and of type I interferon
(IFN-I) in particular in HTLV-1 pathogenesis is still a matter of debate.
ADAR1-p150 is an Interferon Stimulated Gene (ISG) induced by IFN-I that can edit
viral RNAs. We therefore investigated whether it could play the role of an
anti-HTLV factor. Results We demonstrate here that ADAR1 is also expressed in the absence of
IFN stimulation in activated primary T-lymphocytes that are the natural target of
this virus and in HTLV-1 or HTLV-2 chronically infected T-cells. ADAR1 expression
is also increased in primary lymphocytes obtained from HTLV-1 infected
individuals. We show that ADAR1 enhances HTLV-1 and HTLV-2 infection in
T-lymphocytes and that this proviral effect is independent from its editing
activity. ADAR1 expression suppresses IFN-α inhibitory effect on HTLV-1 and HTLV-2
and acts through the repression of PKR phosphorylation. Discussion This study demonstrates that two interferon stimulated genes, i.e.
PKR and ADAR1 have opposite effects on HTLV replication in
vivo. The balanced expression of those proteins could determine the
fate of the viral cycle in the course of infection. Electronic supplementary material The online version of this article (doi:10.1186/s12977-014-0093-9) contains supplementary material, which is available to authorized
users.
Collapse
Affiliation(s)
- Anne Cachat
- Equipe Oncogenèse Rétrovirale, Lyon, Cedex 07, France. .,Equipe labellisée "Ligue Nationale Contre le Cancer", Lyon, Cedex 07, France. .,Centre international de recherche en infectiologie, INSERM U1111 - CNRS UMR5308, Lyon, Cedex 07, France. .,Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364, Lyon, Cedex 07, France. .,Université Lyon 1, LabEx ECOFECT - Eco-evolutionary dynamics of infectious diseases, 69364, Lyon, Cedex 07, France.
| | - Sandrine Alais
- Equipe Oncogenèse Rétrovirale, Lyon, Cedex 07, France. .,Equipe labellisée "Ligue Nationale Contre le Cancer", Lyon, Cedex 07, France. .,Centre international de recherche en infectiologie, INSERM U1111 - CNRS UMR5308, Lyon, Cedex 07, France. .,Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364, Lyon, Cedex 07, France. .,Université Lyon 1, LabEx ECOFECT - Eco-evolutionary dynamics of infectious diseases, 69364, Lyon, Cedex 07, France.
| | - Sébastien Alain Chevalier
- Equipe Oncogenèse Rétrovirale, Lyon, Cedex 07, France. .,Equipe labellisée "Ligue Nationale Contre le Cancer", Lyon, Cedex 07, France. .,Centre international de recherche en infectiologie, INSERM U1111 - CNRS UMR5308, Lyon, Cedex 07, France. .,Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364, Lyon, Cedex 07, France. .,Université Lyon 1, LabEx ECOFECT - Eco-evolutionary dynamics of infectious diseases, 69364, Lyon, Cedex 07, France.
| | - Chloé Journo
- Equipe Oncogenèse Rétrovirale, Lyon, Cedex 07, France. .,Equipe labellisée "Ligue Nationale Contre le Cancer", Lyon, Cedex 07, France. .,Centre international de recherche en infectiologie, INSERM U1111 - CNRS UMR5308, Lyon, Cedex 07, France. .,Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364, Lyon, Cedex 07, France. .,Université Lyon 1, LabEx ECOFECT - Eco-evolutionary dynamics of infectious diseases, 69364, Lyon, Cedex 07, France.
| | - Floriane Fusil
- Centre international de recherche en infectiologie, INSERM U1111 - CNRS UMR5308, Lyon, Cedex 07, France. .,Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364, Lyon, Cedex 07, France. .,Université Lyon 1, LabEx ECOFECT - Eco-evolutionary dynamics of infectious diseases, 69364, Lyon, Cedex 07, France. .,Equipe virus enveloppés, Lyon, Cedex 07, France.
| | - Hélène Dutartre
- Equipe Oncogenèse Rétrovirale, Lyon, Cedex 07, France. .,Equipe labellisée "Ligue Nationale Contre le Cancer", Lyon, Cedex 07, France. .,Centre international de recherche en infectiologie, INSERM U1111 - CNRS UMR5308, Lyon, Cedex 07, France. .,Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364, Lyon, Cedex 07, France. .,Université Lyon 1, LabEx ECOFECT - Eco-evolutionary dynamics of infectious diseases, 69364, Lyon, Cedex 07, France.
| | - Adrien Boniface
- Equipe Oncogenèse Rétrovirale, Lyon, Cedex 07, France. .,Equipe labellisée "Ligue Nationale Contre le Cancer", Lyon, Cedex 07, France. .,Centre international de recherche en infectiologie, INSERM U1111 - CNRS UMR5308, Lyon, Cedex 07, France. .,Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364, Lyon, Cedex 07, France. .,Université Lyon 1, LabEx ECOFECT - Eco-evolutionary dynamics of infectious diseases, 69364, Lyon, Cedex 07, France. .,Biology Department, Master Biosciences, Lyon, Cedex 07, France.
| | - Nga Ling Ko
- Unité d'épidémiologie et physiopathoglogie des virus oncogènes, Institut Pasteur, Paris, 75015, France.
| | - Antoine Gessain
- Unité d'épidémiologie et physiopathoglogie des virus oncogènes, Institut Pasteur, Paris, 75015, France.
| | - François-Loïc Cosset
- Centre international de recherche en infectiologie, INSERM U1111 - CNRS UMR5308, Lyon, Cedex 07, France. .,Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364, Lyon, Cedex 07, France. .,Université Lyon 1, LabEx ECOFECT - Eco-evolutionary dynamics of infectious diseases, 69364, Lyon, Cedex 07, France. .,Equipe virus enveloppés, Lyon, Cedex 07, France.
| | - Rodolphe Suspène
- Unité de rétrovirologie moléculaire, Institut Pasteur, Paris, 75015, France.
| | | | - Renaud Mahieux
- Equipe Oncogenèse Rétrovirale, Lyon, Cedex 07, France. .,Equipe labellisée "Ligue Nationale Contre le Cancer", Lyon, Cedex 07, France. .,Centre international de recherche en infectiologie, INSERM U1111 - CNRS UMR5308, Lyon, Cedex 07, France. .,Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364, Lyon, Cedex 07, France. .,Université Lyon 1, LabEx ECOFECT - Eco-evolutionary dynamics of infectious diseases, 69364, Lyon, Cedex 07, France.
| |
Collapse
|
36
|
Inosine-mediated modulation of RNA sensing by Toll-like receptor 7 (TLR7) and TLR8. J Virol 2013; 88:799-810. [PMID: 24227841 DOI: 10.1128/jvi.01571-13] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
RNA-specific adenosine deaminase (ADAR)-mediated adenosine-to-inosine (A-to-I) editing is a critical arm of the antiviral response. However, mechanistic insights into how A-to-I RNA editing affects viral infection are lacking. We posited that inosine incorporation into RNA facilitates sensing of nonself RNA by innate immune sensors and accordingly investigated the impact of inosine-modified RNA on Toll-like receptor 7 and 8 (TLR7/8) sensing. Inosine incorporation into synthetic single-stranded RNA (ssRNA) potentiated tumor necrosis factor alpha (TNF-α) or alpha interferon (IFN-α) production in human peripheral blood mononuclear cells (PBMCs) in a sequence-dependent manner, indicative of TLR7/8 recruitment. The effect of inosine incorporation on TLR7/8 sensing was restricted to immunostimulatory ssRNAs and was not seen with inosine-containing short double-stranded RNAs or with a deoxy-inosine-modified ssRNA. Inosine-mediated increase of self-secondary structure of an ssRNA resulted in potentiated IFN-α production in human PBMCs through TLR7 recruitment, as established through the use of a TLR7 antagonist and Tlr7-deficient cells. There was a correlation between hyperediting of influenza A viral ssRNA and its ability to stimulate TNF-α, independent of 5'-triphosphate residues, and involving Adar-1. Furthermore, A-to-I editing of viral ssRNA directly enhanced mouse Tlr7 sensing, when present in proportions reproducing biologically relevant levels of RNA editing. Thus, we demonstrate for the first time that inosine incorporation into immunostimulatory ssRNA can potentiate TLR7/8 activation. Our results suggest a novel function of A-to-I RNA editing, which is to facilitate TLR7/8 sensing of phagocytosed viral RNA.
Collapse
|
37
|
T-705 (favipiravir) induces lethal mutagenesis in influenza A H1N1 viruses in vitro. J Virol 2013; 87:3741-51. [PMID: 23325689 DOI: 10.1128/jvi.02346-12] [Citation(s) in RCA: 288] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Several novel anti-influenza compounds are in various phases of clinical development. One of these, T-705 (favipiravir), has a mechanism of action that is not fully understood but is suggested to target influenza virus RNA-dependent RNA polymerase. We investigated the mechanism of T-705 activity against influenza A (H1N1) viruses by applying selective drug pressure over multiple sequential passages in MDCK cells. We found that T-705 treatment did not select specific mutations in potential target proteins, including PB1, PB2, PA, and NP. Phenotypic assays based on cell viability confirmed that no T-705-resistant variants were selected. In the presence of T-705, titers of infectious virus decreased significantly (P < 0.0001) during serial passage in MDCK cells inoculated with seasonal influenza A (H1N1) viruses at a low multiplicity of infection (MOI; 0.0001 PFU/cell) or with 2009 pandemic H1N1 viruses at a high MOI (10 PFU/cell). There was no corresponding decrease in the number of viral RNA copies; therefore, specific virus infectivity (the ratio of infectious virus yield to viral RNA copy number) was reduced. Sequence analysis showed enrichment of G→A and C→T transversion mutations, increased mutation frequency, and a shift of the nucleotide profiles of individual NP gene clones under drug selection pressure. Our results demonstrate that T-705 induces a high rate of mutation that generates a nonviable viral phenotype and that lethal mutagenesis is a key antiviral mechanism of T-705. Our findings also explain the broad spectrum of activity of T-705 against viruses of multiple families.
Collapse
|
38
|
Abstract
Adenosine deaminase acting on RNA (ADAR) catalyzes the hydrolytic deamination of adenosine to inosine in double-stranded RNA (dsRNA) substrates. Inosine pairs preferentially with cytidine, as opposed to uridine; therefore, ADAR editing alters the sequence and base pairing properties of both protein-coding and non-coding RNA. Editing can directly alter the sequence of protein-coding transcripts and modify splicing, or affect a variety of non-coding targets, including microRNA, small interfering RNA, viral transcripts, and repeat elements such as Alu and LINE. Such editing has a wide range of physiological effects, including modification of targets in the brain and in disease states.
Collapse
Affiliation(s)
- Arka Mallela
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
39
|
Wulff BE, Nishikura K. Substitutional A-to-I RNA editing. WILEY INTERDISCIPLINARY REVIEWS-RNA 2012; 1:90-101. [PMID: 21072321 DOI: 10.1002/wrna.10] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Adenosine-to-inosine (A-to-I) editing catalyzed by adenosine deaminases acting on RNA (ADARs) entails the chemical conversion of adenosine residues to inosine residues within double-stranded RNA (dsRNA) substrates. Inosine base pairs as guanosine and A-to-I editing can therefore alter the structure and base pairing properties of the RNA molecule. This has a biological significance in controlling the amount of functional RNA molecules in the cell, in expanding the functionality of a limited set of transcripts, and in defending the cell against certain RNA viruses. A-to-I editing is not limited to any specific type of RNA substrate. Instead, it can affect any RNA molecule able to attain the required double-stranded structure. This includes microRNAs, small interfering RNAs, viral RNAs, and messenger RNAs with potential for recoding events and splice site modifications.
Collapse
Affiliation(s)
- Bjorn-Erik Wulff
- Gene Expression and Regulation, The Wistar Institute, Philadelphia, PA, USA.
| | | |
Collapse
|
40
|
Abstract
Double-stranded RNA (dsRNA) functions both as a substrate of ADARs and also as a molecular trigger of innate immune responses. ADARs, adenosine deaminases that act on RNA, catalyze the deamination of adenosine (A) to produce inosine (I) in dsRNA. ADARs thereby can destablize RNA structures, because the generated I:U mismatch pairs are less stable than A:U base pairs. Additionally, I is read as G instead of A by ribosomes during translation and by viral RNA-dependent RNA polymerases during RNA replication. Members of several virus families have the capacity to produce dsRNA during viral genome transcription and replication. Sequence changes (A-G, and U-C) characteristic of A-I editing can occur during virus growth and persistence. Foreign viral dsRNA also mediates both the induction and the action of interferons. In this chapter our current understanding of the role and significance of ADARs in the context of innate immunity, and as determinants of the outcome of viral infection, will be considered.
Collapse
Affiliation(s)
- Charles E Samuel
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
41
|
Liao JY, Thakur SA, Zalinger ZB, Gerrish KE, Imani F. Inosine-containing RNA is a novel innate immune recognition element and reduces RSV infection. PLoS One 2011; 6:e26463. [PMID: 22028885 PMCID: PMC3196583 DOI: 10.1371/journal.pone.0026463] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 09/27/2011] [Indexed: 12/24/2022] Open
Abstract
During viral infections, single- and double-stranded RNA (ssRNA and dsRNA) are recognized by the host and induce innate immune responses. The cellular enzyme ADAR-1 (adenosine deaminase acting on RNA-1) activation in virally infected cells leads to presence of inosine-containing RNA (Ino-RNA). Here we report that ss-Ino-RNA is a novel viral recognition element. We synthesized unmodified ssRNA and ssRNA that had 6% to16% inosine residues. The results showed that in primary human cells, or in mice, 10% ss-Ino-RNA rapidly and potently induced a significant increase in inflammatory cytokines, such as interferon (IFN)-β (35 fold), tumor necrosis factor (TNF)-α (9.7 fold), and interleukin (IL)-6 (11.3 fold) (p<0.01). Flow cytometry data revealed a corresponding 4-fold increase in influx of neutrophils into the lungs by ss-Ino-RNA treatment. In our in vitro experiments, treatment of epithelial cells with ss-Ino-RNA reduced replication of respiratory syncytial virus (RSV). Interestingly, RNA structural analysis showed that ss-Ino-RNA had increased formation of secondary structures. Our data further revealed that extracellular ss-Ino-RNA was taken up by scavenger receptor class-A (SR-A) which activated downstream MAP Kinase pathways through Toll-like receptor 3 (TLR3) and dsRNA-activated protein kinase (PKR). Our data suggests that ss-Ino-RNA is an as yet undescribed virus-associated innate immune stimulus.
Collapse
Affiliation(s)
- Jie-ying Liao
- Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, United States of America
| | - Sheetal A. Thakur
- Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, United States of America
| | - Zachary B. Zalinger
- Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, United States of America
| | - Kevin E. Gerrish
- Gene Array Core Facility, National Institute of Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, United States of America
| | - Farhad Imani
- ViraSource Laboratories, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
42
|
Enhancement of replication of RNA viruses by ADAR1 via RNA editing and inhibition of RNA-activated protein kinase. J Virol 2011; 85:8460-6. [PMID: 21490091 DOI: 10.1128/jvi.00240-11] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Adenosine deaminase acting on RNA 1 (ADAR1) is a double-stranded RNA binding protein and RNA-editing enzyme that modifies cellular and viral RNAs, including coding and noncoding RNAs. This interferon (IFN)-induced protein was expected to have an antiviral role, but recent studies have demonstrated that it promotes the replication of many RNA viruses. The data from these experiments show that ADAR1 directly enhances replication of hepatitis delta virus, human immunodeficiency virus type 1, vesicular stomatitis virus, and measles virus. The proviral activity of ADAR1 occurs through two mechanisms: RNA editing and inhibition of RNA-activated protein kinase (PKR). While these pathways have been found independently, the two mechanisms can act in concert to increase viral replication and contribute to viral pathogenesis. This novel type of proviral regulation by an IFN-induced protein, combined with some antiviral effects of hyperediting, sheds new light on the importance of ADAR1 during viral infection and transforms our overall understanding of the innate immune response.
Collapse
|
43
|
Lofts LL, Wells JB, Bavari S, Warfield KL. Key genomic changes necessary for an in vivo lethal mouse marburgvirus variant selection process. J Virol 2011; 85:3905-17. [PMID: 21289122 PMCID: PMC3126133 DOI: 10.1128/jvi.02372-10] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 01/18/2011] [Indexed: 11/20/2022] Open
Abstract
Marburgvirus (MARV) infections are generally lethal in humans and nonhuman primates but require in vivo lethal mouse variant selection by the serial transfer (passage) of the nonlethal virus into naïve mice to propagate a lethal infection. The passage of progenitor (wild-type) MARV or Ravn virus (RAVV) from infected scid BALB/c mouse liver homogenates into immunocompetent BALB/c mice results in the selection of lethal mouse viruses from within the quasispecies sufficient to establish lethality in immunocompetent mice. Genomic analysis in conjunction with the passage history of each mutation detailed the altered primary and secondary structures of the viral genomic RNA throughout the process. Key findings included the following: (i) a VP40:D184N mutation previously identified in the lethal guinea pig MARV genome was the first mutation to occur during the passage of both the MARV and RAVV variants; (ii) there was biased hypermutagenesis in the RAVV variant genome; (iii) there were two identical mutations in lethal mouse MARV and RAVV variants, VP40:Y19H in the PPPY motif and VP40:D184N in a loop structure between the two VP40 domains; (iv) the passage of wild-type MARV and RAVV in mice resulted in the selection of viral variants from among the quasispecies with different genotypes than those of the wild-type viruses; and (v) a lethal mouse RAVV variant had different tissue tropisms distinct from those of its wild-type virus. These studies provide insights into how marburgviruses manipulate the host for enzymes, metabolites, translation regulators, and effectors of the innate immune response to serve as potential viral countermeasures.
Collapse
Affiliation(s)
- Loreen L Lofts
- USAMRIID, Virology Division, 1425 Porter Street, Frederick, MD 21702, USA.
| | | | | | | |
Collapse
|
44
|
Samuel CE. Adenosine deaminases acting on RNA (ADARs) are both antiviral and proviral. Virology 2011; 411:180-93. [PMID: 21211811 DOI: 10.1016/j.virol.2010.12.004] [Citation(s) in RCA: 245] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 12/04/2010] [Indexed: 12/18/2022]
Abstract
A-to-I RNA editing, the deamination of adenosine (A) to inosine (I) that occurs in regions of RNA with double-stranded character, is catalyzed by a family of Adenosine Deaminases Acting on RNA (ADARs). In mammals there are three ADAR genes. Two encode proteins that possess demonstrated deaminase activity: ADAR1, which is interferon-inducible, and ADAR2 which is constitutively expressed. ADAR3, by contrast, has not yet been shown to be an active enzyme. The specificity of the ADAR1 and ADAR2 deaminases ranges from highly site-selective to non-selective, dependent on the duplex structure of the substrate RNA. A-to-I editing is a form of nucleotide substitution editing, because I is decoded as guanosine (G) instead of A by ribosomes during translation and by polymerases during RNA-dependent RNA replication. Additionally, A-to-I editing can alter RNA structure stability as I:U mismatches are less stable than A:U base pairs. Both viral and cellular RNAs are edited by ADARs. A-to-I editing is of broad physiologic significance. Among the outcomes of A-to-I editing are biochemical changes that affect how viruses interact with their hosts, changes that can lead to either enhanced or reduced virus growth and persistence depending upon the specific virus.
Collapse
Affiliation(s)
- Charles E Samuel
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
45
|
Ward SV, George CX, Welch MJ, Liou LY, Hahm B, Lewicki H, de la Torre JC, Samuel CE, Oldstone MB. RNA editing enzyme adenosine deaminase is a restriction factor for controlling measles virus replication that also is required for embryogenesis. Proc Natl Acad Sci U S A 2011; 108:331-6. [PMID: 21173229 PMCID: PMC3017198 DOI: 10.1073/pnas.1017241108] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Measles virus (MV), a member of the family Paramyxoviridae and an exclusively human pathogen, is among the most infectious viruses. A progressive fatal neurodegenerative complication, subacute sclerosing panencephalitis (SSPE), occurs during persistent MV infection of the CNS and is associated with biased hypermutations of the viral genome. The observed hypermutations of A-to-G are consistent with conversions catalyzed by the adenosine deaminase acting on RNA (ADAR1). To evaluate the role of ADAR1 in MV infection, we selectively disrupted expression of the IFN-inducible p150 ADAR1 isoform and found it caused embryonic lethality at embryo day (E) 11-E12. We therefore generated p150-deficient and WT mouse embryo fibroblast (MEF) cells stably expressing the MV receptor signaling lymphocyte activation molecule (SLAM or CD150). The p150(-/-) but not WT MEF cells displayed extensive syncytium formation and cytopathic effect (CPE) following infection with MV, consistent with an anti-MV role of the p150 isoform of ADAR1. MV titers were 3 to 4 log higher in p150(-/-) cells compared with WT cells at 21 h postinfection, and restoration of ADAR1 in p150(-/-) cells prevented MV cytopathology. In contrast to infection with MV, p150 disruption had no effect on vesicular stomatitis virus, reovirus, or lymphocytic choriomeningitis virus replication but protected against CPE resulting from infection with Newcastle disease virus, Sendai virus, canine distemper virus, and influenza A virus. Thus, ADAR1 is a restriction factor in the replication of paramyxoviruses and orthomyxoviruses.
Collapse
Affiliation(s)
- Simone V. Ward
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Cyril X. George
- Department of Molecular, Cellular, and Developmental Biology and
| | - Megan J. Welch
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Li-Ying Liou
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Bumsuk Hahm
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
- Departments of Surgery and Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65212
| | - Hanna Lewicki
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Juan C. de la Torre
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| | - Charles E. Samuel
- Department of Molecular, Cellular, and Developmental Biology and
- Biomolecular Sciences and Engineering Program, University of California, Santa Barbara, CA 92106; and
| | - Michael B. Oldstone
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
46
|
George CX, Gan Z, Liu Y, Samuel CE. Adenosine deaminases acting on RNA, RNA editing, and interferon action. J Interferon Cytokine Res 2010; 31:99-117. [PMID: 21182352 DOI: 10.1089/jir.2010.0097] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Adenosine deaminases acting on RNA (ADARs) catalyze adenosine (A) to inosine (I) editing of RNA that possesses double-stranded (ds) structure. A-to-I RNA editing results in nucleotide substitution, because I is recognized as G instead of A both by ribosomes and by RNA polymerases. A-to-I substitution can also cause dsRNA destabilization, as I:U mismatch base pairs are less stable than A:U base pairs. Three mammalian ADAR genes are known, of which two encode active deaminases (ADAR1 and ADAR2). Alternative promoters together with alternative splicing give rise to two protein size forms of ADAR1: an interferon-inducible ADAR1-p150 deaminase that binds dsRNA and Z-DNA, and a constitutively expressed ADAR1-p110 deaminase. ADAR2, like ADAR1-p110, is constitutively expressed and binds dsRNA. A-to-I editing occurs with both viral and cellular RNAs, and affects a broad range of biological processes. These include virus growth and persistence, apoptosis and embryogenesis, neurotransmitter receptor and ion channel function, pancreatic cell function, and post-transcriptional gene regulation by microRNAs. Biochemical processes that provide a framework for understanding the physiologic changes following ADAR-catalyzed A-to-I ( = G) editing events include mRNA translation by changing codons and hence the amino acid sequence of proteins; pre-mRNA splicing by altering splice site recognition sequences; RNA stability by changing sequences involved in nuclease recognition; genetic stability in the case of RNA virus genomes by changing sequences during viral RNA replication; and RNA-structure-dependent activities such as microRNA production or targeting or protein-RNA interactions.
Collapse
Affiliation(s)
- Cyril X George
- Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, California 93106, USA
| | | | | | | |
Collapse
|
47
|
Double-stranded RNA adenosine deaminase ADAR-1-induced hypermutated genomes among inactivated seasonal influenza and live attenuated measles virus vaccines. J Virol 2010; 85:2458-62. [PMID: 21159878 DOI: 10.1128/jvi.02138-10] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We sought to examine ADAR-1 editing of measles and influenza virus genomes derived from inactivated seasonal influenza and live attenuated measles virus vaccines grown on chicken cells as the culture substrate. Using highly sensitive 3DI-PCR (R. Suspène et al., Nucleic Acids Res. 36:e72, 2008), it was possible to show that ADAR-1 could hyperdeaminate adenosine residues in both measles virus and influenza virus A genomes. Detailed analysis of the dinucleotide editing context showed preferences for 5'ArA and 5'UrA, which is typical of editing in mammalian cells. The hyperedited mutant frequency, including genomes and antigenomes, was a log greater for influenza virus compared to measles virus, suggesting a greater sensitivity to restriction by ADAR-1.
Collapse
|
48
|
Borrow P, Martínez-Sobrido L, de la Torre JC. Inhibition of the type I interferon antiviral response during arenavirus infection. Viruses 2010; 2:2443-80. [PMID: 21994626 PMCID: PMC3185579 DOI: 10.3390/v2112443] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 10/22/2010] [Accepted: 10/22/2010] [Indexed: 12/20/2022] Open
Abstract
Arenaviruses merit interest both as tractable experimental model systems to study acute and persistent viral infections, and as clinically-important human pathogens. Several arenaviruses cause hemorrhagic fever (HF) disease in humans. In addition, evidence indicates that the globally-distributed prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) is a human pathogen of clinical significance in congenital infections, and also poses a great danger to immunosuppressed individuals. Arenavirus persistence and pathogenesis are facilitated by their ability to overcome the host innate immune response. Mammalian hosts have developed both membrane toll-like receptors (TLR) and cytoplasmic pattern recognition receptors (PRRs) that recognize specific pathogen-associated molecular patterns (PAMPs), resulting in activation of the transcription factors IRF3 or IRF7, or both, which together with NF-κB and ATF-2/c-JUN induce production of type I interferon (IFN-I). IFN-I plays a key role in host anti-microbial defense by mediating direct antiviral effects via up-regulation of IFN-I stimulated genes (ISGs), activating dendritic cells (DCs) and natural killer (NK) cells, and promoting the induction of adaptive responses. Accordingly, viruses have developed a plethora of strategies to disrupt the IFN-I mediated antiviral defenses of the host, and the viral gene products responsible for these disruptions are often major virulence determinants. IRF3- and IRF7-dependent induction of host innate immune responses is frequently targeted by viruses. Thus, the arenavirus nucleoprotein (NP) was shown to inhibit the IFN-I response by interfering with the activation of IRF3. This NP anti-IFN activity, together with alterations in the number and function of DCs observed in mice chronically infected with LCMV, likely play an important role in LCMV persistence in its murine host. In this review we will discuss current knowledge about the cellular and molecular mechanisms by which arenaviruses can subvert the host innate immune response and their implications for understanding HF arenaviral disease as well as arenavirus persistence in their natural hosts.
Collapse
Affiliation(s)
- Persephone Borrow
- Nuffield Department of Clinical Medicine, The Jenner Institute, University of Oxford, Compton, Newbury, Berkshire RG20 7NN, UK; E-Mail:
| | - Luis Martínez-Sobrido
- Department of Microbiology and Immunology, University of Rochester, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Juan Carlos de la Torre
- Department of Immunology and Microbial Science, IMM-6, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| |
Collapse
|
49
|
Martín V, Abia D, Domingo E, Grande-Pérez A. An interfering activity against lymphocytic choriomeningitis virus replication associated with enhanced mutagenesis. J Gen Virol 2009; 91:990-1003. [PMID: 20007356 DOI: 10.1099/vir.0.017053-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Previous studies have documented that, in the presence of the mutagenic base analogue 5-fluorouracil (FU), lymphocytic choriomeningitis virus (LCMV) that persisted in BHK-21 cells decreased its infectivity to a larger extent than intracellular viral RNA levels, prior to virus extinction. This observation, together with in silico simulations, led to the proposal of the lethal defection model of virus extinction. This model suggests the participation of defective-interfering genomes in the loss of infectivity by increased mutagenesis. Since LCMV naturally produces defective-interfering particles, it was important to show that a capacity to interfere is produced in association with FU treatment. Here, we document that BHK-21 cells persistently infected with LCMV grown in the presence of FU, but not in its absence, generated an interfering activity that suppressed LCMV infectivity. Interference was specific for LCMV and was sensitive to UV irradiation and its activity was dose- and time-dependent. The interfering preparations produced positive LCMV immunofluorescence and viral particles seen by electron microscopy when used to infect cells, despite some preparations being devoid of detectable infectivity. Interference did not involve significant increases of mutant spectrum complexity, as predicted by the lethal defection model. The results provide support for a specific interference associated with LCMV when the virus replicates in the presence of FU. The excess of interference relative to that observed in the absence of FU is necessary for LCMV extinction.
Collapse
Affiliation(s)
- Verónica Martín
- Centro de Biología Molecular 'Severo Ochoa' (CSIC-UAM), C/ Nicolás Cabrera, 1, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | | | | | | |
Collapse
|
50
|
Cai L, Li Y, Liu F, Zhang W, Huo B, Zheng W, Ding R, Guo J, Zhao Q, Dou K. The influence of ADAR1’s regulation on lymphocyte cell function during rejection. Mol Biol Rep 2009; 37:2703-9. [DOI: 10.1007/s11033-009-9804-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 09/02/2009] [Indexed: 12/14/2022]
|