2016
DOI: 10.1038/nmicrobiol.2016.11
|View full text |Cite
|
Sign up to set email alerts
|

Dynamics of the human and viral m6A RNA methylomes during HIV-1 infection of T cells

Abstract: N6-methyladenosine (m6A) is the most prevalent internal modification of eukaryotic mRNA. Very little is known of the function of m6A in the immune system or its role in host–pathogen interactions. Here we investigated the topology, dynamics, and bidirectional influences of the viral–host RNA methylomes during HIV-1 infection of human CD4 T cells. We show that viral infection triggers a massive increase in m6A in both host and viral mRNAs. In HIV-1 mRNA, we identified 14 methylation peaks in coding and noncodin… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
2

Citation Types

31
560
3
1

Year Published

2017
2017
2022
2022

Publication Types

Select...
5
2

Relationship

0
7

Authors

Journals

citations
Cited by 398 publications
(614 citation statements)
references
References 47 publications
31
560
3
1
Order By: Relevance
“…2), one might anticipate that, if viral RNAs are indeed m 6 A modified, this would primarily or exclusively occur for RNAs generated by nuclear DNA or RNA viruses. In fact, analyses of three DNA viruses (adenovirus, herpes simplex virus type 1, and SV40), four retroviruses (the closely related avian sarcoma virus and Rous sarcoma virus as well as HIV-1 and feline leukemia virus), and the orthomyxovirus influenza A virus (IAV) have revealed m 6 A residues present at levels that are at least as high as the number of m 6 A residues detected on cellular mRNAs (4)(5)(6)(7)(8)(9)(10)(11)(43)(44)(45)(46). Moreover, in HIV-1, where m 6 A residues have been mapped at near single-nucleotide resolution, the consensus m 6 A addition sites that are utilized are highly conserved across HIV-1 isolates (10).…”
Section: Nuclear Rna and Dna Virusesmentioning
confidence: 99%
See 2 more Smart Citations
“…2), one might anticipate that, if viral RNAs are indeed m 6 A modified, this would primarily or exclusively occur for RNAs generated by nuclear DNA or RNA viruses. In fact, analyses of three DNA viruses (adenovirus, herpes simplex virus type 1, and SV40), four retroviruses (the closely related avian sarcoma virus and Rous sarcoma virus as well as HIV-1 and feline leukemia virus), and the orthomyxovirus influenza A virus (IAV) have revealed m 6 A residues present at levels that are at least as high as the number of m 6 A residues detected on cellular mRNAs (4)(5)(6)(7)(8)(9)(10)(11)(43)(44)(45)(46). Moreover, in HIV-1, where m 6 A residues have been mapped at near single-nucleotide resolution, the consensus m 6 A addition sites that are utilized are highly conserved across HIV-1 isolates (10).…”
Section: Nuclear Rna and Dna Virusesmentioning
confidence: 99%
“…We note that m 6 A addition has been proposed to affect mRNA splicing (24)(25)(26), stability (27,47), and translation (47)(48)(49)(50), to modify RNA structure (51), and to inhibit the recognition of viral RNAs by Toll-like receptors and RIG-I (52, 53), and so m 6 A could positively regulate several aspects of the viral life cycle. Indeed, knockdown of the METTL3 and/or METTL14 m 6 A writers using RNA interference (RNAi) has been reported to inhibit HIV-1 replication up to 5-fold, while knockdown of the ALKBH5 m 6 A demethylase enhanced HIV-1 replication up to 8-fold (11,14). Similarly, in CD4-positive T cells, overexpression 6 A reader proteins YTHDF1, YTHDF2, and YTHDF3 (upper panels) and of the writer components METTL3, METTL14, and WTAP (lower panels), and were then subjected to immunofluorescence using an anti-FLAG antibody.…”
Section: Nuclear Rna and Dna Virusesmentioning
confidence: 99%
See 1 more Smart Citation
“…Reducing methylation at this site by knocking down METTL3/METTL14 or by mutating the site substantially inhibits viral RNA export into the cytoplasm, an essential step in the HIV-1 replication cycle. 50 Interestingly, Tirumuru et al report a binding site for the m 6 A-binding protein YTHDF1 in this region. The proteins YTHDF1, YTHDF2 and YTHDF3, all established m 6 A-binding proteins, appear to negatively regulate HIV-1 viral reverse transcription and mRNA transcription in this study.…”
Section: Viral Infectionmentioning
confidence: 99%
“…[49][50][51] Though each study took different approaches and reaches different conclusions in some cases, they do converge in many respects. They find that HIV-1 RNA carries multiple m 6 A peaks that appear to be important in infection, and that the cellular host responds with infection-specific methylation of multiple transcripts potentially involved in the response to viral infection.…”
Section: Viral Infectionmentioning
confidence: 99%