Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Sci Rep ; 12(1): 14972, 2022 Sep 13.
Article in English | MEDLINE | ID: covidwho-2028722

ABSTRACT

During COVID-19 pandemic, mutations of SARS-CoV-2 produce new strains that can be more infectious or evade vaccines. Viral RNA mutations can arise from misincorporation by RNA-polymerases and modification by host factors. Analysis of SARS-CoV-2 sequence from patients showed a strong bias toward C-to-U mutation, suggesting a potential mutational role by host APOBEC cytosine deaminases that possess broad anti-viral activity. We report the first experimental evidence demonstrating that APOBEC3A, APOBEC1, and APOBEC3G can edit on specific sites of SARS-CoV-2 RNA to produce C-to-U mutations. However, SARS-CoV-2 replication and viral progeny production in Caco-2 cells are not inhibited by the expression of these APOBECs. Instead, expression of wild-type APOBEC3 greatly promotes viral replication/propagation, suggesting that SARS-CoV-2 utilizes the APOBEC-mediated mutations for fitness and evolution. Unlike the random mutations, this study suggests the predictability of all possible viral genome mutations by these APOBECs based on the UC/AC motifs and the viral genomic RNA structure.


Subject(s)
COVID-19 , RNA Editing , APOBEC Deaminases/genetics , APOBEC Deaminases/metabolism , COVID-19/genetics , Caco-2 Cells , Cytidine Deaminase , Humans , Mutation , Pandemics , Proteins , RNA, Viral/genetics , RNA, Viral/metabolism , SARS-CoV-2/genetics
2.
Infect Genet Evol ; 97: 105188, 2022 01.
Article in English | MEDLINE | ID: covidwho-1568934

ABSTRACT

The best and most effective way to combat pandemics is to use effective vaccines and live attenuated vaccines are among the most effective vaccines. However, one of the major problems is the length of time it takes to get the attenuated vaccines. Today, the CRISPR toolkit (Clustered Regularly Inerspaced Short Palindromic Repeats) has made it possible to make changes with high efficiency and speed. Using this toolkit to make point mutations on the RNA virus's genome in a coculture of permissive and nonpermissive cells and under controlled conditions can accelerate changes in the genome and accelerate natural selection to obtain live attenuated vaccines.


Subject(s)
COVID-19 Vaccines/genetics , COVID-19/prevention & control , CRISPR-Cas Systems , Gene Editing/methods , Mutation Rate , SARS-CoV-2/genetics , Viral Proteins/genetics , APOBEC Deaminases/genetics , APOBEC Deaminases/immunology , Adenosine Deaminase/genetics , Adenosine Deaminase/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , COVID-19/immunology , COVID-19 Vaccines/biosynthesis , Endonucleases/genetics , Endonucleases/immunology , Gene Expression , Genome, Viral , Humans , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , SARS-CoV-2/immunology , Selection, Genetic , Vaccines, Attenuated , Viral Proteins/immunology
3.
Brief Bioinform ; 23(1)2022 01 17.
Article in English | MEDLINE | ID: covidwho-1406466

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has triggered an unprecedented international effort to sequence complete viral genomes. We leveraged this wealth of information to characterize the substitution spectrum of SARS-CoV-2 and to compare it with those of other human and animal coronaviruses. We show that, once nucleotide composition is taken into account, human and most animal coronaviruses display a mutation spectrum dominated by C to U and G to U substitutions, a feature that is not shared by other positive-sense RNA viruses. However, the proportions of C to U and G to U substitutions tend to decrease as divergence increases, suggesting that, whatever their origin, a proportion of these changes is subsequently eliminated by purifying selection. Analysis of the sequence context of C to U substitutions showed little evidence of apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC)-mediated editing and such contexts were similar for SARS-CoV-2 and Middle East respiratory syndrome coronavirus sampled from different hosts, despite different repertoires of APOBEC3 proteins in distinct species. Conversely, we found evidence that C to U and G to U changes affect CpG dinucleotides at a frequency higher than expected. Whereas this suggests ongoing selective reduction of CpGs, this effect alone cannot account for the substitution spectra. Finally, we show that, during the first months of SARS-CoV-2 pandemic spread, the frequency of both G to U and C to U substitutions increased. Our data suggest that the substitution spectrum of SARS-CoV-2 is determined by an interplay of factors, including intrinsic biases of the replication process, avoidance of CpG dinucleotides and other constraints exerted by the new host.


Subject(s)
COVID-19/genetics , Evolution, Molecular , Genome, Viral , Mutation , Pandemics , SARS-CoV-2/genetics , APOBEC Deaminases/genetics , APOBEC Deaminases/metabolism , Animals , COVID-19/epidemiology , Humans , Phylogeny , SARS-CoV-2/metabolism
4.
Scand J Immunol ; 94(5): e13100, 2021 Nov.
Article in English | MEDLINE | ID: covidwho-1388399

ABSTRACT

The SARS-CoV-2 epidemic infections in Australia during 2020 were small in number in epidemiological terms and are well described. The SARS-CoV-2 genomic sequence data of many infected patients have been largely curated in a number of publicly available databases, including the corresponding epidemiological data made available by the Victorian Department of Health and Human Services. We have critically analysed the available SARS-CoV-2 haplotypes and genomic sequences in the context of putative deficits in innate immune APOBEC and ADAR deaminase anti-viral responses. It is now known that immune impaired elderly co-morbid patients display clear deficits in interferon type 1 (α/ß) and III (λ) stimulated innate immune gene cascades, of which APOBEC and ADAR induced expression are part. These deficiencies may help explain some of the clear genetic patterns in SARS-CoV-2 genomes isolated in Victoria, Australia, during the 2nd Wave (June-September, 2020). We tested the hypothesis that predicted lowered innate immune APOBEC and ADAR anti-viral deaminase responses in a significant proportion of elderly patients would be consistent with/reflected in a low level of observed mutagenesis in many isolated SARS-CoV-2 genomes. Our findings are consistent with this expectation. The analysis also supports the conclusions of the Victorian government's Department of Health that essentially one variant or haplotype infected Victorian aged care facilities where the great majority (79%) of all 820 SARS-CoV-2 associated deaths occurred. The implications of our data analysis for other localized epidemics and efficient coronavirus vaccine design and delivery are discussed.


Subject(s)
APOBEC Deaminases/genetics , Adenosine Deaminase/genetics , COVID-19 Vaccines/immunology , COVID-19/immunology , RNA-Binding Proteins/genetics , SARS-CoV-2/physiology , APOBEC Deaminases/metabolism , Adenosine Deaminase/metabolism , Age Factors , Aged, 80 and over , COVID-19/epidemiology , COVID-19/virology , Female , Gene Regulatory Networks , Haplotypes , Humans , Immunity, Innate , Immunologic Deficiency Syndromes , Interferon Type I/genetics , Male , RNA-Binding Proteins/metabolism , Victoria/epidemiology
5.
Genes (Basel) ; 12(6)2021 05 27.
Article in English | MEDLINE | ID: covidwho-1256475

ABSTRACT

The genomic diversity of SARS-CoV-2 has been a focus during the ongoing COVID-19 pandemic. Here, we analyzed the distribution and character of emerging mutations in a data set comprising more than 95,000 virus genomes covering eight major SARS-CoV-2 lineages in the GISAID database, including genotypes arising during COVID-19 therapy. Globally, the C>U transitions and G>U transversions were the most represented mutations, accounting for the majority of single-nucleotide variations. Mutational spectra were not influenced by the time the virus had been circulating in its host or medical treatment. At the amino acid level, we observed about a 2-fold excess of substitutions in favor of hydrophobic amino acids over the reverse. However, most mutations constituting variants of interests of the S-protein (spike) lead to hydrophilic amino acids, counteracting the global trend. The C>U and G>U substitutions altered codons towards increased amino acid hydrophobicity values in more than 80% of cases. The bias is explained by the existing differences in the codon composition for amino acids bearing contrasting biochemical properties. Mutation asymmetries apparently influence the biochemical features of SARS CoV-2 proteins, which may impact protein-protein interactions, fusion of viral and cellular membranes, and virion assembly.


Subject(s)
COVID-19/virology , Genome, Viral , Hydrophobic and Hydrophilic Interactions , Mutation , SARS-CoV-2/genetics , Viral Proteins/chemistry , Viral Proteins/genetics , APOBEC Deaminases , Alleles , Amino Acid Substitution , Amino Acids/chemistry , Amino Acids/genetics , Evolution, Molecular , Genetic Variation , Genotype , Host-Pathogen Interactions , Humans , Phylogeny , Polymorphism, Single Nucleotide , Protein Binding , Protein Interaction Domains and Motifs , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics
6.
PLoS Pathog ; 17(6): e1009596, 2021 06.
Article in English | MEDLINE | ID: covidwho-1249581

ABSTRACT

The rapid evolution of RNA viruses has been long considered to result from a combination of high copying error frequencies during RNA replication, short generation times and the consequent extensive fixation of neutral or adaptive changes over short periods. While both the identities and sites of mutations are typically modelled as being random, recent investigations of sequence diversity of SARS coronavirus 2 (SARS-CoV-2) have identified a preponderance of C->U transitions, proposed to be driven by an APOBEC-like RNA editing process. The current study investigated whether this phenomenon could be observed in datasets of other RNA viruses. Using a 5% divergence filter to infer directionality, 18 from 36 datasets of aligned coding region sequences from a diverse range of mammalian RNA viruses (including Picornaviridae, Flaviviridae, Matonaviridae, Caliciviridae and Coronaviridae) showed a >2-fold base composition normalised excess of C->U transitions compared to U->C (range 2.1x-7.5x), with a consistently observed favoured 5' U upstream context. The presence of genome scale RNA secondary structure (GORS) was the only other genomic or structural parameter significantly associated with C->U/U->C transition asymmetries by multivariable analysis (ANOVA), potentially reflecting RNA structure dependence of sites targeted for C->U mutations. Using the association index metric, C->U changes were specifically over-represented at phylogenetically uninformative sites, potentially paralleling extensive homoplasy of this transition reported in SARS-CoV-2. Although mechanisms remain to be functionally characterised, excess C->U substitutions accounted for 11-14% of standing sequence variability of structured viruses and may therefore represent a potent driver of their sequence diversification and longer-term evolution.


Subject(s)
Mammals/virology , Mutation , RNA Viruses/genetics , SARS-CoV-2/genetics , APOBEC Deaminases/metabolism , Animals , Base Sequence , COVID-19/virology , Cytidine/genetics , DNA Damage/physiology , Evolution, Molecular , Gene Expression Regulation, Viral , Genome, Viral , Host-Pathogen Interactions/genetics , Humans , Nucleic Acid Conformation , Phylogeny , RNA Editing/physiology , RNA Viruses/classification , RNA, Viral/chemistry , RNA, Viral/genetics , SARS-CoV-2/chemistry , SARS-CoV-2/classification , Sequence Analysis, RNA , Transcription, Genetic/genetics , Uridine/genetics
7.
Biochem Biophys Res Commun ; 538: 35-39, 2021 01 29.
Article in English | MEDLINE | ID: covidwho-1139448

ABSTRACT

The extensive sequence data generated from SARS-CoV-2 during the 2020 pandemic has facilitated the study of viral genome evolution over a brief period of time. This has highlighted instances of directional mutation pressures exerted on the SARS-CoV-2 genome from host antiviral defense systems. In this brief review we describe three such human defense mechanisms, the apolipoprotein B mRNA editing catalytic polypeptide-like proteins (APOBEC), adenosine deaminase acting on RNA proteins (ADAR), and reactive oxygen species (ROS), and discuss their potential implications on SARS-CoV-2 evolution.


Subject(s)
APOBEC Deaminases/metabolism , Adenosine Deaminase/metabolism , COVID-19/virology , Gene Editing , Genome, Viral , Host-Pathogen Interactions/genetics , RNA-Binding Proteins/metabolism , SARS-CoV-2/genetics , COVID-19/epidemiology , Humans , Reactive Oxygen Species/metabolism
8.
Clin Immunol ; 226: 108699, 2021 05.
Article in English | MEDLINE | ID: covidwho-1101151

ABSTRACT

RNA editing is a fundamental biological process with 2 major forms, namely adenosine-to-inosine (A-to-I, recognized as A-to-G) and cytosine-to-uracil (C-to-U) deamination, mediated by ADAR and APOBEC enzyme families, respectively. A-to-I RNA editing has been shown to directly affect the genome/transcriptome of RNA viruses with significant repercussions for viral protein synthesis, proliferation and infectivity, while it also affects recognition of double-stranded RNAs by cytosolic receptors controlling the host innate immune response. Recent evidence suggests that RNA editing may be present in SARS-CoV-2 genome/transcriptome. The majority of mapped mutations in SARS-CoV-2 genome are A-to-G/U-to-C(opposite strand) and C-to-U/G-to-A(opposite strand) substitutions comprising potential ADAR-/APOBEC-mediated deamination events. A single nucleotide substitution can have dramatic effects on SARS-CoV-2 infectivity as shown by the D614G(A-to-G) substitution in the spike protein. Future studies utilizing serial sampling from patients with COVID-19 are warranted to delineate whether RNA editing affects viral replication and/or the host immune response to SARS-CoV-2.


Subject(s)
APOBEC Deaminases/metabolism , Adenosine Deaminase/metabolism , COVID-19/immunology , Immunity, Innate , RNA Editing , RNA Viruses/genetics , RNA-Binding Proteins/metabolism , SARS-CoV-2/genetics , APOBEC Deaminases/genetics , Adenosine Deaminase/genetics , COVID-19/enzymology , COVID-19/virology , Humans , Mutation , RNA Viruses/pathogenicity , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/genetics , SARS-CoV-2/metabolism
9.
Virology ; 556: 62-72, 2021 04.
Article in English | MEDLINE | ID: covidwho-1065649

ABSTRACT

Members of the APOBEC family of cytidine deaminases show antiviral activities in mammalian cells through lethal editing in the genomes of small DNA viruses, herpesviruses and retroviruses, and potentially those of RNA viruses such as coronaviruses. Consistent with the latter, APOBEC-like directional C→U transitions of genomic plus-strand RNA are greatly overrepresented in SARS-CoV-2 genome sequences of variants emerging during the COVID-19 pandemic. A C→U mutational process may leave evolutionary imprints on coronavirus genomes, including extensive homoplasy from editing and reversion at targeted sites and the occurrence of driven amino acid sequence changes in viral proteins. If sustained over longer periods, this process may account for the previously reported marked global depletion of C and excess of U bases in human seasonal coronavirus genomes. This review synthesizes the current knowledge on APOBEC evolution and function and the evidence of their role in APOBEC-mediated genome editing of SARS-CoV-2 and other coronaviruses.


Subject(s)
APOBEC Deaminases/metabolism , Coronavirus/genetics , Evolution, Molecular , Genome, Viral/genetics , RNA Editing , APOBEC Deaminases/chemistry , APOBEC Deaminases/genetics , Animals , Coronavirus Infections/virology , Humans , Mutation , SARS-CoV-2/genetics
11.
Virologie (Montrouge) ; 24(6): 381-418, 2020 12 01.
Article in French | MEDLINE | ID: covidwho-1030336

ABSTRACT

The innate immune response is nonspecific and constitutes the first line of defense against infections by pathogens, mainly by enabling their elimination by phagocytosis or apoptosis. In immune cells, this response is characterized, amongst others, by the synthesis of restriction factors, a class of proteins whose role is to inhibit viral replication. Among them, the proteins of the APOBEC3 (Apolipoprotein B mRNA-editing Enzyme Catalytic polypeptide-like 3 or A3) family are major antiviral factors that target a wide range of viruses. One of their targets is the Human Immunodeficiency Virus Type 1 (HIV-1): the deaminase activity of some A3 proteins converts a fraction of cytidines of the viral genome into uridines, impairing its expression. Nevertheless, HIV-1 counteracts A3 proteins thanks to its Vif protein, which inhibits them by hijacking several cellular mechanisms. Besides, APOBEC3 proteins help maintaining the genome integrity by inhibiting retroelements but they also contribute to carcinogenesis, as it is the case for A3A and A3B, two major factors in this process. The large range of A3 activities, combined with recent studies showing their implication in the regulation of emerging viruses (Zika, SARS-CoV-2), allow A3 and their viral partners to be considered as therapeutic areas.


Subject(s)
APOBEC Deaminases/physiology , COVID-19/immunology , Immunity, Innate , Adult , Amino Acid Motifs , Animals , Cell Cycle Proteins/metabolism , Cytidine Deaminase/physiology , DNA Repair , DNA, Viral/metabolism , Deamination , Humans , Mammals/metabolism , MicroRNAs/genetics , Models, Molecular , Molecular Targeted Therapy , Mutagenesis , Neoplasms/enzymology , Neoplasms/etiology , Neoplasms/genetics , Prognosis , Protein Conformation , RNA Editing , Structure-Activity Relationship , Transcription, Genetic , Viral Proteins/metabolism , Virus Diseases/drug therapy , Virus Diseases/enzymology , Virus Diseases/immunology , Virus Replication
12.
PLoS One ; 15(12): e0244025, 2020.
Article in English | MEDLINE | ID: covidwho-992706

ABSTRACT

Coronaviruses such as SARS-CoV-2 regularly infect host tissues that express antiviral proteins (AVPs) in abundance. Understanding how they evolve to adapt or evade host immune responses is important in the effort to control the spread of infection. Two AVPs that may shape viral genomes are the zinc finger antiviral protein (ZAP) and the apolipoprotein B mRNA editing enzyme-catalytic polypeptide-like 3 (APOBEC3). The former binds to CpG dinucleotides to facilitate the degradation of viral transcripts while the latter frequently deaminates C into U residues which could generate notable viral sequence variations. We tested the hypothesis that both APOBEC3 and ZAP impose selective pressures that shape the genome of an infecting coronavirus. Our investigation considered a comprehensive number of publicly available genomes for seven coronaviruses (SARS-CoV-2, SARS-CoV, and MERS infecting Homo sapiens, Bovine CoV infecting Bos taurus, MHV infecting Mus musculus, HEV infecting Sus scrofa, and CRCoV infecting Canis lupus familiaris). We show that coronaviruses that regularly infect tissues with abundant AVPs have CpG-deficient and U-rich genomes; whereas those that do not infect tissues with abundant AVPs do not share these sequence hallmarks. Among the coronaviruses surveyed herein, CpG is most deficient in SARS-CoV-2 and a temporal analysis showed a marked increase in C to U mutations over four months of SARS-CoV-2 genome evolution. Furthermore, the preferred motifs in which these C to U mutations occur are the same as those subjected to APOBEC3 editing in HIV-1. These results suggest that both ZAP and APOBEC3 shape the SARS-CoV-2 genome: ZAP imposes a strong CpG avoidance, and APOBEC3 constantly edits C to U. Evolutionary pressures exerted by host immune systems onto viral genomes may motivate novel strategies for SARS-CoV-2 vaccine development.


Subject(s)
COVID-19/genetics , Coronavirus/genetics , Cytidine Deaminase/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , APOBEC Deaminases , Animals , COVID-19/pathology , COVID-19/virology , Cattle , Coronavirus/classification , Coronavirus/pathogenicity , Dogs , Evolution, Molecular , Genome, Viral/genetics , Humans , Mice , Middle East Respiratory Syndrome Coronavirus/genetics , Middle East Respiratory Syndrome Coronavirus/pathogenicity , Severe acute respiratory syndrome-related coronavirus/genetics , Severe acute respiratory syndrome-related coronavirus/pathogenicity , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Swine/virology
13.
Sci Rep ; 10(1): 17766, 2020 10 20.
Article in English | MEDLINE | ID: covidwho-882928

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.


Subject(s)
Betacoronavirus/immunology , Coronavirus Infections/pathology , Pneumonia, Viral/pathology , APOBEC Deaminases/metabolism , Adenosine Deaminase/metabolism , Betacoronavirus/classification , Betacoronavirus/genetics , Betacoronavirus/isolation & purification , COVID-19 , Cell Line, Tumor , Coronavirus Infections/immunology , Coronavirus Infections/virology , Humans , Interleukin-6/metabolism , Pandemics , Phylogeny , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , Point Mutation , RNA Editing , SARS-CoV-2 , Tumor Necrosis Factor-alpha/metabolism , Uracil/metabolism
14.
PLoS Pathog ; 16(8): e1008718, 2020 08.
Article in English | MEDLINE | ID: covidwho-717612

ABSTRACT

APOBEC3 enzymes are innate immune effectors that introduce mutations into viral genomes. These enzymes are cytidine deaminases which transform cytosine into uracil. They preferentially mutate cytidine preceded by thymidine making the 5'TC motif their favored target. Viruses have evolved different strategies to evade APOBEC3 restriction. Certain viruses actively encode viral proteins antagonizing the APOBEC3s, others passively face the APOBEC3 selection pressure thanks to a depleted genome for APOBEC3-targeted motifs. Hence, the APOBEC3s left on the genome of certain viruses an evolutionary footprint. The aim of our study is the identification of these viruses having a genome shaped by the APOBEC3s. We analyzed the genome of 33,400 human viruses for the depletion of APOBEC3-favored motifs. We demonstrate that the APOBEC3 selection pressure impacts at least 22% of all currently annotated human viral species. The papillomaviridae and polyomaviridae are the most intensively footprinted families; evidencing a selection pressure acting genome-wide and on both strands. Members of the parvoviridae family are differentially targeted in term of both magnitude and localization of the footprint. Interestingly, a massive APOBEC3 footprint is present on both strands of the B19 erythroparvovirus; making this viral genome one of the most cleaned sequences for APOBEC3-favored motifs. We also identified the endemic coronaviridae as significantly footprinted. Interestingly, no such footprint has been detected on the zoonotic MERS-CoV, SARS-CoV-1 and SARS-CoV-2 coronaviruses. In addition to viruses that are footprinted genome-wide, certain viruses are footprinted only on very short sections of their genome. That is the case for the gamma-herpesviridae and adenoviridae where the footprint is localized on the lytic origins of replication. A mild footprint can also be detected on the negative strand of the reverse transcribing HIV-1, HIV-2, HTLV-1 and HBV viruses. Together, our data illustrate the extent of the APOBEC3 selection pressure on the human viruses and identify new putatively APOBEC3-targeted viruses.


Subject(s)
Cytidine Deaminase/metabolism , Genome, Viral/genetics , Host-Pathogen Interactions/genetics , Selection, Genetic/genetics , Virus Replication/genetics , APOBEC Deaminases , Coronaviridae/genetics , Humans , Immunity, Innate/immunology , Papillomaviridae/genetics , Parvoviridae/genetics , Polyomaviridae/genetics , Viral Proteins/genetics
15.
mSphere ; 5(3)2020 06 24.
Article in English | MEDLINE | ID: covidwho-612518

ABSTRACT

The pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has motivated an intensive analysis of its molecular epidemiology following its worldwide spread. To understand the early evolutionary events following its emergence, a data set of 985 complete SARS-CoV-2 sequences was assembled. Variants showed a mean of 5.5 to 9.5 nucleotide differences from each other, consistent with a midrange coronavirus substitution rate of 3 × 10-4 substitutions/site/year. Almost one-half of sequence changes were C→U transitions, with an 8-fold base frequency normalized directional asymmetry between C→U and U→C substitutions. Elevated ratios were observed in other recently emerged coronaviruses (SARS-CoV, Middle East respiratory syndrome [MERS]-CoV), and decreasing ratios were observed in other human coronaviruses (HCoV-NL63, -OC43, -229E, and -HKU1) proportionate to their increasing divergence. C→U transitions underpinned almost one-half of the amino acid differences between SARS-CoV-2 variants and occurred preferentially in both 5' U/A and 3' U/A flanking sequence contexts comparable to favored motifs of human APOBEC3 proteins. Marked base asymmetries observed in nonpandemic human coronaviruses (U ≫ A > G ≫ C) and low G+C contents may represent long-term effects of prolonged C→U hypermutation in their hosts. The evidence that much of sequence change in SARS-CoV-2 and other coronaviruses may be driven by a host APOBEC-like editing process has profound implications for understanding their short- and long-term evolution. Repeated cycles of mutation and reversion in favored mutational hot spots and the widespread occurrence of amino acid changes with no adaptive value for the virus represent a quite different paradigm of virus sequence change from neutral and Darwinian evolutionary frameworks and are not incorporated by standard models used in molecular epidemiology investigations.IMPORTANCE The wealth of accurately curated sequence data for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), its long genome, and its low substitution rate provides a relatively blank canvas with which to investigate effects of mutational and editing processes imposed by the host cell. The finding that a large proportion of sequence change in SARS-CoV-2 in the initial months of the pandemic comprised C→U mutations in a host APOBEC-like context provides evidence for a potent host-driven antiviral editing mechanism against coronaviruses more often associated with antiretroviral defense. In evolutionary terms, the contribution of biased, convergent, and context-dependent mutations to sequence change in SARS-CoV-2 is substantial, and these processes are not incorporated by standard models used in molecular epidemiology investigations.


Subject(s)
Betacoronavirus/genetics , Cytosine/analysis , Genome, Viral/genetics , Polymorphism, Single Nucleotide/genetics , Uracil/analysis , APOBEC Deaminases , Base Composition/genetics , Base Sequence/genetics , COVID-19 , Coronavirus Infections/pathology , Cytidine Deaminase/genetics , Humans , Middle East Respiratory Syndrome Coronavirus/genetics , Pandemics , Pneumonia, Viral/pathology , Severe acute respiratory syndrome-related coronavirus/genetics , SARS-CoV-2
16.
Sci Adv ; 6(25): eabb5813, 2020 06.
Article in English | MEDLINE | ID: covidwho-619103

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.


Subject(s)
Betacoronavirus/genetics , Betacoronavirus/metabolism , Coronavirus Infections/genetics , Host-Pathogen Interactions/genetics , Pneumonia, Viral/genetics , RNA Editing/genetics , Transcriptome , APOBEC Deaminases/genetics , APOBEC Deaminases/metabolism , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Base Sequence/genetics , Bronchoalveolar Lavage Fluid/virology , COVID-19 , Coronavirus Infections/virology , Genome, Viral/genetics , Humans , Mutation Rate , Nucleotides/genetics , Nucleotides/metabolism , Pandemics , Pneumonia, Viral/virology , RNA, Viral/genetics , SARS-CoV-2 , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL