AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (3.3 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Full Length Article | Open Access

Targeting m6A modification inhibits herpes virus 1 infection

Zhuoying FengaFanghang ZhouaMiaomiao TanaTingting WangaYing ChenaWenwen XubBin LibXin Wanga( )Xin Denga( )Ming-Liang Hea,c( )
Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, PR China
MOE Key Laboratory of Tumor Molecular Biology and Guangdong Provincial Key Laboratory of Bioengineering Medicine, National Engineering Research Center of Genetic Medicine, Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong 510632, PR China
CityU Shenzhen Research Institute, Shenzhen, Guangdong 518057, PR China

Peer review under responsibility of Chongqing Medical University.

Show Author Information

Abstract

The latent infection by herpes virus type 1 (HSV-1) may be lifelong in trigeminal ganglia and a suspected cause of Alzheimer's Disease (AD) and Amyotrophic lateral sclerosis (ALS). Whether and how N6-methyladenosine (m6A) modification of viral RNAs affects virus infection are poorly understood. Here, we report that HSV-1 infection enhanced the expression of m6A writers (METTL3, METTL14) and readers (YTHDF1/2/3) at the early infection stage and decreased their expression later on, while suppressed the erasers' (FTO, ALBKH5) expression immediately upon infection to facilitate viral replication. Inhibiting m6A modification by 3-deazaadenosine (DAA) significantly decreased viral replication and reduced viral reproduction over 1000 folds. More interestingly, depleting the writers and readers by siRNAs inhibited virus replication and reproduction; whereas depleting the erasers promoted viral replication and reproduction. Silencing YTHDF3 strikingly decreased viral replication by up to 90%, leading to reduction of up to 10-fold viral replication and over 100-fold virus reproduction, respectively. Depletion of m6A initiator METTL3 (by 60%–70%) by siRNA correlatedly decreased viral replication 60%–70%, and reduced virus yield over 30-fold. Consistently, ectopic expression of METTL3 largely increased virus yield. METTL3 knockdown suppressed the HSV-1 intermediate early and early genes (ICP0, ICP8 and UL23) and late genes (VP16, UL44, UL49 and ICP47); while ectopic expression of METTL3 upregulated these gene expression. Results from our study shed the lights on the importance for m6A modification to initiate HSV-1 early replication. The components of m6A modification machinery, particularly m6A initiator METTL3 and reader YTHDF3, would be potential important targets for combating HSV-1 infections.

References

1

Desrosiers R, Friderici K, Rottman F. Identification of methylated nucleosides in messenger RNA from Novikoff hepatoma cells. Proc Nat Acad Sci U S A. 1974;71(10):3971-3975.

2

Wei CM, Gershowitz A, Moss B. Methylated nucleotides block 5′ terminus of HeLa cell messenger RNA. Cell. 1975;4(4):379-386.

3

Wei CM, Moss B. Nucleotide sequences at the N6-methyladenosine sites of HeLa cell messenger ribonucleic acid. Biochemistry. 1977;16(8):1672-1676.

4

Wang X, Lu Z, Gomez A, et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014;505(7481):117-120.

5

Wei W, Ji X, Guo X, Ji S. Regulatory role of N 6 -methyladenosine (m 6 A) methylation in RNA processing and human diseases. J Cell Biochem. 2017;118(9):2534-2543.

6

Wei CM, Gershowitz A, Moss B. 5′-Terminal and internal methylated nucleotide sequences in HeLa cell mRNA. Biochemistry. 1976;15(2):397-401.

7

Batista PJ, Molinie B, Wang J, et al. m(6)A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell. 2014;15(6):707-719.

8

Dominissini D, Moshitch-Moshkovitz S, Schwartz S, et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature. 2012;485(7397):201-206.

9

Huang J, Yin P. Structural insights into N6-methyladenosine (m6A) modification in the transcriptome. Genomics Proteomics Bioinformatics. 2018;16(2):85-89.

10

Wang X, Huang J, Zou T, Yin P. Human m(6)A writers: two subunits, 2 roles. RNA Biol. 2017;14(3):300-304.

11

Zhang C, Fu J, Zhou Y. A review in research progress concerning m6A methylation and immunoregulation. Front Immunol. 2019;10:922.

12

Wang X, Zhao BS, Roundtree IA, et al. N6-methyladenosine modulates messenger RNA translation efficiency. Cell. 2015;161(6):1388-1399.

13

Roignant JY, Soller M. m6A in mRNA: an ancient mechanism for fine-tuning gene expression. Trends Genet. 2017;33(6):380-390.

14

Shi H, Wang X, Lu Z, et al. YTHDF3 facilitates translation and decay of N(6)-methyladenosine-modified RNA. Cell Res. 2017;27(3):315-328.

15

Zheng G, Dahl JA, Niu Y, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013;49(1):18-29.

16

Jia G, Fu Y, Zhao X, et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol. 2011;7(12):885-887.

17

Fu Y, Jia G, Pang X, et al. FTO-mediated formation of N6-hydroxymethyladenosine and N6-formyladenosine in mammalian RNA. Nat Commun. 2013;4:1798.

18

Batista PJ, Molinie B, Wang J, et al. m(6)A RNA modification controls cell fate transition in mammalian embryonic stem cells. Cell Stem Cell. 2014;15(6):707-719.

19

Widagdo J, Anggono V. The m6A-epitranscriptomic signature in neurobiology: from neurodevelopment to brain plasticity. J Neurochem. 2018;147(2):137-152.

20

Chen J, Fang X, Zhong P, Song Z, Hu X. N6-methyladenosine modifications: interactions with novel RNA-binding proteins and roles in signal transduction. RNA Biol. 2019;16(8):991-1000.

21

Wu DF, Koch T, Liang YJ, et al. Membrane glycoprotein M6a interacts with the micro-opioid receptor and facilitates receptor endocytosis and recycling. J Biol Chem. 2007;282(30):22239-22247.

22

Sekar D, Lakshmanan G. Methylation of N6-adenosine (m6A) modification in miRNAs and its implications in immunity. Epigenomics. 2020;12(13):1083-1085.

23

Han M, Liu Z, Xu Y, et al. Abnormality of m6A mRNA methylation is involved in Alzheimer’s disease. Front Neurosci. 2020;14:98.

24

Wang S, Sun C, Li J, et al. Roles of RNA methylation by means of N(6)-methyladenosine (m(6)A) in human cancers. Cancer Lett. 2017;408:112-120.

25

Brocard M, Ruggieri A, Locker N. m6A RNA methylation, a new hallmark in virus-host interactions. J Gen Virol. 2017;98(9):2207-2214.

26

McGeoch DJ, Rixon FJ, Davison AJ. Topics in herpesvirus genomics and evolution. Virus Res. 2006;117(1):90-104.

27

Amin I, Vajeeha A, Younas S, et al. HSV-1 infection: role of viral proteins and cellular receptors. Crit Rev Eukaryot Gene Expr. 2019;29(5):461-469.

28

Wan Q, Song D, Li H, He ML. Stress proteins: the biological functions in virus infection, present and challenges for target-based antiviral drug development. Sign Transduct Target Ther. 2020;5(1):125.

29

Danastas K, Miranda-Saksena M, Cunningham AL. Herpes simplex virus type 1 interactions with the interferon system. Int J Mol Sci. 2020;21(14):5150.

30

Dimock K, Stoltzfus CM. Sequence specificity of internal methylation in B77 avian sarcoma virus RNA subunits. Biochemistry. 1977;16(3):471-478.

31

Hashimoto SI, Green M. Multiple methylated cap sequences in adenovirus type 2 early mRNA. J Virol. 1976;20(2):425-435.

32

Kim GW, Imam H, Khan M, Siddiqui A. N6-Methyladenosine modification of hepatitis B and C viral RNAs attenuates host innate immunity via RIG-I signaling. J Biol Chem. 2020;295(37):13123-13133.

33

Zhou F, Wan Q, Lu J, Chen Y, Lu G, He ML. Pim1 impacts enterovirus A71 replication and represents a potential target in antiviral therapy. iScience. 2019;19:715-727.

34

Schremser V, Antoniewicz L, Tschachler E, Geusau A. Polymerase chain reaction for the diagnosis of herpesvirus infections in dermatology : analysis of clinical data. Wien Klin Wochenschr. 2020;132(1-2):35-41.

35

Dan X, Wan Q, Yi L, et al. Hsp27 responds to and facilitates enterovirus A71 replication by enhancing viral internal ribosome entry site-mediated translation. J Virol. 2019;93(9):e02322–18.

36

Dominissini D, Moshitch-Moshkovitz S, Salmon-Divon M, Amariglio N, Rechavi G. Transcriptome-wide mapping of N(6)-methyladenosine by m(6)A-seq based on immunocapturing and massively parallel sequencing. Nat Protoc. 2013;8(1):176-189.

37

Meng J, Lu Z, Liu H, et al. A protocol for RNA methylation differential analysis with MeRIP-Seq data and exomePeak R/Bioconductor package. Methods. 2014;69(3):274-281.

38

Imam H, Khan M, Gokhale NS. N6-methyladenosine modification of hepatitis B virus RNA differentially regulates the viral life cycle. Proc Nat Acad Sci U S A. 2018;115(35):8829-8834.

39

Gokhale NS, Mclntyre ABR, McFadden MJ, et al. N6-Methyladenosine in flaviviridae viral RNA genomes regulates infection. Cell Host Microbe. 2016;20(5):654-665.

40

de Clercq E, Montgomery JA. Broad-spectrum antiviral activity of the carbocyclic analog of 3-deazaadenosine. Antivir Res. 1983;3(1):17-24.

41

Montgomery JA, Clayton SJ, Thomas HJ. Carbocyclic analog of 3-deazaadenosine - a novel antiviral agent using S-adenosylhomocysteine hydrolase as a pharmacological target. J Med Chem. 1982;25(6):626-629.

42

Xiang S, Liang X, Yin S, Liu J, Xiang Z. N6-methyladenosine methyltransferase METTL3 promotes colorectal cancer cell proliferation through enhancing MYC expression. Am J Transl Res. 2020;12(5):1789-1806.

43

Zhou L, Yang C, Zhang N, Zhang X, Zhao T, Yu J. Silencing METTL3 inhibits the proliferation and invasion of osteosarcoma by regulating ATAD2. Biomed Pharmacother. 2020;125:109964.

44

Komala Sari T, Gianopulos KA, Nicola AV. Glycoprotein C of herpes simplex virus 1 shields glycoprotein B from antibody neutralization. J Virol. 2020;94(5):e01852-19.

45

Cheng JT, Wang YY, Zhu LZ, et al. Novel transcription regulatory sequences and factors of the immune evasion protein ICP47 (US12) of herpes simplex viruses. Virol J. 2020;17(1):101.

46

Goldsmith K, Chen W, Johnson DC, Hendricks RL. Infected cell protein (ICP)47 enhances herpes simplex virus neurovirulence by blocking the CD8+ T cell response. J Exp Med. 1998;187(3):341-348.

47

Huang CY, Yao HW, Wang LC, Shen FH, Hsu SM, Chen SH. Thymidine kinase-negative herpes simplex virus 1 can efficiently establish persistent infection in neural tissues of nude mice. J Virol. 2017;91(4):e01979–16.

48

Burrel S, Deback C, Agut H, Boutolleau D. Genotypic characterization of UL23 thymidine kinase and UL30 DNA polymerase of clinical isolates of herpes simplex virus: natural polymorphism and mutations associated with resistance to antivirals. Antimicrob Agents Chemother. 2010;54(11):4833-4842.

49

Hao H, Hao S, Chen H. N6-methyladenosine modification and METTL3 modulate enterovirus 71 replication. Nucleic Acids Res. 2019;47(1):362-374.

50

Kennedy EM, Bogerd HP, Kornepati AV, et al. Posttranscriptional m6A editing of HIV-1 mRNAs enhances viral gene expression. Cell Host Microbe. 2016;19(5):675-685.

51

Klupp BG, Baumeister J, Karger A, Visser N, Mettenleiter TC. Identification and characterization of a novel structural glycoprotein in pseudorabies virus, gL. J Virol. 1994;68(6):3868-3878.

52

Reynolds AE, Ryckman BJ, Baines JD, Zhou Y, Liang L, Roller RJ. U(L)31 and U(L)34 proteins of herpes simplex virus type 1 form a complex that accumulates at the nuclear rim and is required for envelopment of nucleocapsids. J Virol. 2001;75(18):8803-8817.

53

Hernández Durán A, Greco TM, Vollmer B, Cristea IM, Grünewald K, Topf M. Protein interactions and consensus clustering analysis uncover insights into herpesvirus virion structure and function relationships. PLoS Biol. 2019;17(6):e3000316.

54

You H, Zheng S, Huang Z, Lin Y, Shen Q, Zheng C. Herpes simplex virus 1 tegument protein UL46 inhibits TANK-binding kinase 1-mediated signaling. mBio. 2019;10(3):e00919-19.

55

Elliott GD. The extreme carboxyl terminus of the equine herpesvirus 1 homolog of herpes simplex virus VP16 is essential for immediate-early gene activation. J Virol. 1994;68(8):4890-4897.

56

Tsai K, Courtney DG, Cullen BR. Addition of m6A to SV40 late mRNAs enhances viral structural gene expression and replication. PLoS Pathog. 2018;14(2):e1006919.

57

Kennedy EM, Bogerd HP, Kornepati AV, et al. Posttranscriptional m(6)A editing of HIV-1 mRNAs enhances viral gene expression. Cell Host Microbe. 2016;19(5):675-685.

58

Tirumuru N, Zhao BS, Lu W, Lu Z, He C, Wu L. Correction: N(6)-methyladenosine of HIV-1 RNA regulates viral infection and HIV-1 Gag protein expression. Elife. 2017;6:e31482.

59

Lichinchi G, Zhao BS, Wu Y, et al. Dynamics of human and viral RNA methylation during Zika virus infection. Cell Host Microbe. 2016;20(5):666-673.

Genes & Diseases
Pages 1114-1128
Cite this article:
Feng Z, Zhou F, Tan M, et al. Targeting m6A modification inhibits herpes virus 1 infection. Genes & Diseases, 2022, 9(4): 1114-1128. https://doi.org/10.1016/j.gendis.2021.02.004

318

Views

9

Downloads

28

Crossref

26

Web of Science

28

Scopus

0

CSCD

Altmetrics

Received: 30 September 2020
Revised: 26 January 2021
Accepted: 10 February 2021
Published: 22 February 2021
© 2021, Chongqing Medical University.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return