N6-Methyladenosine in Flaviviridae Viral RNA Genomes Regulates Infection

Summary The RNA modification N6-methyladenosine (m6A) post-transcriptionally regulates RNA function. The cellular machinery that controls m6A includes methyltransferases and demethylases that add or remove this modification, as well as m6A-binding YTHDF proteins that promote the translation or degradation of m6A-modified mRNA. We demonstrate that m6A modulates infection by hepatitis C virus (HCV). Depletion of m6A methyltransferases or an m6A demethylase, respectively, increases or decreases infectious HCV particle production. During HCV infection, YTHDF proteins relocalize to lipid droplets, sites of viral assembly, and their depletion increases infectious viral particles. We further mapped m6A sites across the HCV genome and determined that inactivating m6A in one viral genomic region increases viral titer without affecting RNA replication. Additional mapping of m6A on the RNA genomes of other Flaviviridae, including dengue, Zika, yellow fever, and West Nile virus, identifies conserved regions modified by m6A. Altogether, this work identifies m6A as a conserved regulatory mark across Flaviviridae genomes.

[1]  Saeed Tavazoie,et al.  HNRNPA2B1 Is a Mediator of m6A-Dependent Nuclear RNA Processing Events , 2015, Cell.

[2]  P. Sarnow,et al.  Modulation of Hepatitis C Virus RNA Abundance by a Liver-Specific MicroRNA , 2005, Science.

[3]  Chuan He,et al.  N6-methyladenosine of HIV-1 RNA regulates viral infection and HIV-1 Gag protein expression , 2016, eLife.

[4]  Zhike Lu,et al.  m6A-dependent regulation of messenger RNA stability , 2013, Nature.

[5]  Samie R. Jaffrey,et al.  The dynamic epitranscriptome: N6-methyladenosine and gene expression control , 2014, Nature Reviews Molecular Cell Biology.

[6]  M. Diamond,et al.  A Viral RNA Structural Element Alters Host Recognition of Nonself RNA , 2014, Science.

[7]  Cara T. Pager,et al.  Modulation of hepatitis C virus RNA abundance and virus release by dispersion of processing bodies and enrichment of stress granules. , 2013, Virology.

[8]  B. Semler,et al.  Viral Determinants of miR-122-Independent Hepatitis C Virus Replication , 2015, mSphere.

[9]  C. Rice,et al.  CD81 Is Required for Hepatitis C Virus Glycoprotein-Mediated Viral Infection , 2004, Journal of Virology.

[10]  Johannes E. Schindelin,et al.  Fiji: an open-source platform for biological-image analysis , 2012, Nature Methods.

[11]  Chuan He,et al.  RNA N6-methyladenosine methylation in post-transcriptional gene expression regulation , 2015, Genes & development.

[12]  S. Hopcraft,et al.  Selection of a hepatitis C virus with altered entry factor requirements reveals a genetic interaction between the E1 glycoprotein and claudins , 2015, Hepatology.

[13]  Michael Gale,et al.  IPS-1 Is Essential for the Control of West Nile Virus Infection and Immunity , 2010, PLoS pathogens.

[14]  S. Akira,et al.  Regulation of innate antiviral defenses through a shared repressor domain in RIG-I and LGP2 , 2007, Proceedings of the National Academy of Sciences.

[15]  Christopher T. Jones,et al.  A Human monoclonal antibody targeting scavenger receptor class B type I precludes hepatitis C virus infection and viral spread in vitro and in vivo , 2012, Hepatology.

[16]  J. Darnell,et al.  The methylation of adenovirus-specific nuclear and cytoplasmic RNA. , 1976, Nucleic acids research.

[17]  M. Garcia-Blanco,et al.  G Protein-Coupled Receptor Kinase 2 Promotes Flaviviridae Entry and Replication , 2012, PLoS neglected tropical diseases.

[18]  Gideon Rechavi,et al.  Transcriptome-wide mapping of N6-methyladenosine by m6A-seq based on immunocapturing and massively parallel sequencing , 2013, Nature Protocols.

[19]  R. Gregory,et al.  The m(6)A Methyltransferase METTL3 Promotes Translation in Human Cancer Cells. , 2016, Molecular cell.

[20]  D. Tyrrell,et al.  IFITM1 is a tight junction protein that inhibits hepatitis C virus entry†, ‡ , 2012, Hepatology.

[21]  Gideon Rechavi,et al.  Gene expression regulation mediated through reversible m6A RNA methylation , 2014, Nature Reviews Genetics.

[22]  Houping Ni,et al.  Suppression of RNA recognition by Toll-like receptors: the impact of nucleoside modification and the evolutionary origin of RNA. , 2005, Immunity.

[23]  K. Dimock,et al.  Sequence specificity of internal methylation in B77 avian sarcoma virus RNA subunits. , 1977, Biochemistry.

[24]  S. Kane,et al.  Inhibition of methylation at two internal N6-methyladenosine sites caused by GAC to GAU mutations. , 1987, The Journal of biological chemistry.

[25]  Shu-Bing Qian,et al.  Dynamic m6A mRNA methylation directs translational control of heat shock response , 2015, Nature.

[26]  C. Romero-Lopez,et al.  The cis-acting replication element of the Hepatitis C virus genome recruits host factors that influence viral replication and translation , 2016, Scientific Reports.

[27]  Shang Gao,et al.  Dynamics of the human and viral m6A RNA methylomes during HIV-1 infection of T cells , 2016, Nature Microbiology.

[28]  O. Elemento,et al.  Comprehensive Analysis of mRNA Methylation Reveals Enrichment in 3′ UTRs and near Stop Codons , 2012, Cell.

[29]  Schraga Schwartz,et al.  Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5' sites. , 2014, Cell reports.

[30]  M. Garcia-Blanco,et al.  Flaviviral RNAs: weapons and targets in the war between virus and host. , 2014, The Biochemical journal.

[31]  Christopher E. Mason,et al.  Single-nucleotide resolution mapping of m6A and m6Am throughout the transcriptome , 2015, Nature Methods.

[32]  Y. Matsuura,et al.  Interaction of Hepatitis C Virus Core Protein with Viral Sense RNA and Suppression of Its Translation , 1999, Journal of Virology.

[33]  Chengqi Yi,et al.  N6-Methyladenosine in Nuclear RNA is a Major Substrate of the Obesity-Associated FTO , 2011, Nature chemical biology.

[34]  Olivier Elemento,et al.  5′ UTR m6A Promotes Cap-Independent Translation , 2015, Cell.

[35]  Gideon Rechavi,et al.  The dynamic N1-methyladenosine methylome in eukaryotic messenger RNA , 2016, Nature.

[36]  James E. DiCarlo,et al.  RNA-Guided Human Genome Engineering via Cas9 , 2013, Science.

[37]  Bryan R. Cullen,et al.  Posttranscriptional m(6)A Editing of HIV-1 mRNAs Enhances Viral Gene Expression. , 2016, Cell host & microbe.

[38]  A. Gamarnik,et al.  Dengue Virus RNA Structure Specialization Facilitates Host Adaptation , 2015, PLoS pathogens.

[39]  Pauline E. Chugh,et al.  Regulation of the hepatitis C virus RNA replicase by endogenous lipid peroxidation , 2014, Nature Medicine.

[40]  S. Temple,et al.  Attomole quantification and global profile of RNA modifications: Epitranscriptome of human neural stem cells , 2015, Nucleic acids research.

[41]  R. Bartenschlager,et al.  The lipid droplet is an important organelle for hepatitis C virus production , 2007, Nature Cell Biology.

[42]  M. Garcia-Blanco,et al.  Discovery of Insect and Human Dengue Virus Host Factors , 2009, Nature.

[43]  G. Saade,et al.  Zika virus: History, emergence, biology, and prospects for control. , 2016, Antiviral research.

[44]  Mounavya Aligeti,et al.  Cooperation between the Hepatitis C Virus p7 and NS5B Proteins Enhances Virion Infectivity , 2015, Journal of Virology.

[45]  R J Roberts,et al.  Sequence specificity of the human mRNA N6-adenosine methylase in vitro. , 1990, Nucleic acids research.

[46]  Cole Trapnell,et al.  Ultrafast and memory-efficient alignment of short DNA sequences to the human genome , 2009, Genome Biology.

[47]  Arne Klungland,et al.  ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. , 2013, Molecular cell.

[48]  B. Pulendran,et al.  Zika Virus Infects Human Placental Macrophages. , 2016, Cell host & microbe.

[49]  R. Krug,et al.  Influenza viral mRNA contains internal N6-methyladenosine and 5'-terminal 7-methylguanosine in cap structures , 1976, Journal of virology.

[50]  Uwe Ohler,et al.  PARalyzer: definition of RNA binding sites from PAR-CLIP short-read sequence data , 2011, Genome Biology.

[51]  F. Chisari,et al.  Differential Biophysical Properties of Infectious Intracellular and Secreted Hepatitis C Virus Particles , 2006, Journal of Virology.

[52]  Miao Yu,et al.  A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation , 2013, Nature chemical biology.

[53]  Robert B. Tesh,et al.  Genetic Characterization of Zika Virus Strains: Geographic Expansion of the Asian Lineage , 2012, PLoS neglected tropical diseases.

[54]  M. Gale,et al.  Alpha Interferon Induces Distinct Translational Control Programs To Suppress Hepatitis C Virus RNA Replication , 2003, Journal of Virology.

[55]  Dali Han,et al.  Modulates Messenger RNA Translation Efficiency , 2015 .

[56]  Qi Zhou,et al.  m(6)A RNA methylation is regulated by microRNAs and promotes reprogramming to pluripotency. , 2015, Cell stem cell.

[57]  M. Kupiec,et al.  Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq , 2012, Nature.

[58]  K. Weeks,et al.  Functionally conserved architecture of hepatitis C virus RNA genomes , 2015, Proceedings of the National Academy of Sciences.

[59]  S. Kane,et al.  Precise localization of m6A in Rous sarcoma virus RNA reveals clustering of methylation sites: implications for RNA processing , 1985, Molecular and cellular biology.

[60]  A. Shatkin,et al.  Methylated simian virus 40-specific RNA from nuclei and cytoplasm of infected BSC-1 cells. , 1975, Proceedings of the National Academy of Sciences of the United States of America.

[61]  Roy M. Anderson,et al.  Underwhelming the Immune Response: Effect of Slow Virus Growth on CD8+-T-Lymphocyte Responses , 2004, Journal of Virology.

[62]  Jonas Korlach,et al.  The birth of the Epitranscriptome: deciphering the function of RNA modifications , 2012, Genome Biology.

[63]  Andrew S. Kohlway,et al.  The Coding Region of the HCV Genome Contains a Network of Regulatory RNA Structures. , 2016, Molecular cell.

[64]  N. Kato,et al.  Hepatitis C Virus Hijacks P-Body and Stress Granule Components around Lipid Droplets , 2011, Journal of Virology.

[65]  Scott B. Dewell,et al.  Transcriptome-wide Identification of RNA-Binding Protein and MicroRNA Target Sites by PAR-CLIP , 2010, Cell.

[66]  H. Erfle,et al.  Identification of HNRNPK as Regulator of Hepatitis C Virus Particle Production , 2015, PLoS pathogens.

[67]  P. Thibault,et al.  A Host YB-1 Ribonucleoprotein Complex Is Hijacked by Hepatitis C Virus for the Control of NS3-Dependent Particle Production , 2013, Journal of Virology.

[68]  Sheng Li,et al.  The pivotal regulatory landscape of RNA modifications. , 2014, Annual review of genomics and human genetics.

[69]  Bing Ren,et al.  N6-methyladenosine-dependent regulation of messenger RNA stability , 2013 .