Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Cell ; 183(5): 1185-1201.e20, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33242417

ABSTRACT

Spaceflight is known to impose changes on human physiology with unknown molecular etiologies. To reveal these causes, we used a multi-omics, systems biology analytical approach using biomedical profiles from fifty-nine astronauts and data from NASA's GeneLab derived from hundreds of samples flown in space to determine transcriptomic, proteomic, metabolomic, and epigenetic responses to spaceflight. Overall pathway analyses on the multi-omics datasets showed significant enrichment for mitochondrial processes, as well as innate immunity, chronic inflammation, cell cycle, circadian rhythm, and olfactory functions. Importantly, NASA's Twin Study provided a platform to confirm several of our principal findings. Evidence of altered mitochondrial function and DNA damage was also found in the urine and blood metabolic data compiled from the astronaut cohort and NASA Twin Study data, indicating mitochondrial stress as a consistent phenotype of spaceflight.


Subject(s)
Genomics , Mitochondria/pathology , Space Flight , Stress, Physiological , Animals , Circadian Rhythm , Extracellular Matrix/metabolism , Humans , Immunity, Innate , Lipid Metabolism , Metabolic Flux Analysis , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscles/immunology , Organ Specificity , Smell/physiology
2.
Mol Cell ; 83(22): 3948-3949, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37977114

ABSTRACT

In this issue, Tapescu et al.1 identify DDX39A as a novel antiviral protein that acts on conserved features of alphavirus RNA to limit infection in an IFN-independent manner.


Subject(s)
Chikungunya virus , Chikungunya virus/genetics , Virus Replication/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Antiviral Agents/pharmacology
3.
Mol Cell ; 77(3): 542-555.e8, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31810760

ABSTRACT

The RNA modification N6-methyladenosine (m6A) modulates mRNA fate and thus affects many biological processes. We analyzed m6A across the transcriptome following infection by dengue virus (DENV), Zika virus (ZIKV), West Nile virus (WNV), and hepatitis C virus (HCV). We found that infection by these viruses in the Flaviviridae family alters m6A modification of specific cellular transcripts, including RIOK3 and CIRBP. During viral infection, the addition of m6A to RIOK3 promotes its translation, while loss of m6A in CIRBP promotes alternative splicing. Importantly, viral activation of innate immune sensing or the endoplasmic reticulum (ER) stress response contributes to the changes in m6A in RIOK3 or CIRBP, respectively. Further, several transcripts with infection-altered m6A profiles, including RIOK3 and CIRBP, encode proteins that influence DENV, ZIKV, and HCV infection. Overall, this work reveals that cellular signaling pathways activated during viral infection lead to alterations in m6A modification of host mRNAs to regulate infection.


Subject(s)
Adenosine/analogs & derivatives , Flaviviridae Infections/genetics , RNA, Messenger/genetics , Adenosine/genetics , Cell Line , Dengue/virology , Dengue Virus/genetics , Flaviviridae/genetics , Hepacivirus/genetics , Hepatitis C/virology , Host-Pathogen Interactions/genetics , Humans , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Virus Replication/genetics , Zika Virus/genetics , Zika Virus Infection/genetics
4.
RNA ; 30(5): 482-490, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38531643

ABSTRACT

Viral RNA molecules contain multiple layers of regulatory information. This includes features beyond the primary sequence, such as RNA structures and RNA modifications, including N6-methyladenosine (m6A). Many recent studies have identified the presence and location of m6A in viral RNA and have found diverse regulatory roles for this modification during viral infection. However, to date, viral m6A mapping strategies have limitations that prevent a complete understanding of the function of m6A on individual viral RNA molecules. While m6A sites have been profiled on bulk RNA from many viruses, the resulting m6A maps of viral RNAs described to date present a composite picture of m6A across viral RNA molecules in the infected cell. Thus, for most viruses, it is unknown if unique viral m6A profiles exist throughout infection, nor if they regulate specific viral life cycle stages. Here, we describe several challenges to defining the function of m6A in viral RNA molecules and provide a framework for future studies to help in the understanding of how m6A regulates viral infection.


Subject(s)
Virus Diseases , Viruses , Humans , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Replication/genetics , RNA/genetics , Viruses/genetics
5.
Nat Immunol ; 15(1): 72-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24241692

ABSTRACT

IFNL3, which encodes interferon-λ3 (IFN-λ3), has received considerable attention in the hepatitis C virus (HCV) field, as many independent genome-wide association studies have identified a strong association between polymorphisms near IFNL3 and clearance of HCV. However, the mechanism underlying this association has remained elusive. In this study, we report the identification of a functional polymorphism (rs4803217) in the 3' untranslated region (UTR) of IFNL3 mRNA that dictated transcript stability. We found that this polymorphism influenced AU-rich element (ARE)-mediated decay (AMD) of IFNL3 mRNA, as well as the binding of HCV-induced microRNAs during infection. Together these pathways mediated robust repression of the unfavorable IFNL3 polymorphism. Our data reveal a previously unknown mechanism by which HCV attenuates the antiviral response and indicate new potential therapeutic targets for HCV treatment.


Subject(s)
AU Rich Elements/genetics , Interleukins/genetics , MicroRNAs/genetics , Polymorphism, Single Nucleotide , RNA Stability/genetics , 3' Untranslated Regions/genetics , Base Sequence , Cell Line, Tumor , Flow Cytometry , Genotype , Hep G2 Cells , Hepacivirus/physiology , Hepatitis C/genetics , Hepatitis C/virology , Host-Pathogen Interactions , Humans , Interferons , Interleukins/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/virology , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid
6.
Proc Natl Acad Sci U S A ; 120(15): e2217053120, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37011200

ABSTRACT

The genomes of RNA viruses encode the information required for replication in host cells both in their linear sequence and in complex higher-order structures. A subset of these RNA genome structures show clear sequence conservation, and have been extensively described for well-characterized viruses. However, the extent to which viral RNA genomes contain functional structural elements-unable to be detected by sequence alone-that nonetheless are critical to viral fitness is largely unknown. Here, we devise a structure-first experimental strategy and use it to identify 22 structure-similar motifs across the coding sequences of the RNA genomes for the four dengue virus serotypes. At least 10 of these motifs modulate viral fitness, revealing a significant unnoticed extent of RNA structure-mediated regulation within viral coding sequences. These viral RNA structures promote a compact global genome architecture, interact with proteins, and regulate the viral replication cycle. These motifs are also thus constrained at the levels of both RNA structure and protein sequence and are potential resistance-refractory targets for antivirals and live-attenuated vaccines. Structure-first identification of conserved RNA structure enables efficient discovery of pervasive RNA-mediated regulation in viral genomes and, likely, other cellular RNAs.


Subject(s)
Dengue , RNA Viruses , Humans , Nucleic Acid Conformation , RNA, Viral/metabolism , RNA Viruses/genetics , Genome, Viral/genetics , Virus Replication/genetics
7.
Trends Biochem Sci ; 46(5): 366-377, 2021 05.
Article in English | MEDLINE | ID: mdl-33309325

ABSTRACT

Recent discoveries have revealed that, during viral infection, the presence of the RNA modification N6-methyladenosine (m6A) on viral and cellular RNAs has profound impacts on infection outcome. Although m6A directly regulates many viral RNA processes, its effects on cellular RNAs and pathways during infection have only recently begun to be elucidated. Disentangling the effects of m6A on viral and host RNAs remains a challenge for the field. m6A has been found to regulate host responses such as viral RNA sensing, cytokine responses, and immune cell functions. We highlight recent findings describing how m6A modulates host responses to viral infection and discuss future directions that will lead to a synergistic understanding of the processes by which m6A regulates viral infection.


Subject(s)
Virus Diseases , Adenosine/analogs & derivatives , Cytokines , Humans , Immunity, Innate , RNA, Viral
8.
J Virol ; 98(1): e0179123, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38168672

ABSTRACT

In the United States (US), biosafety and biosecurity oversight of research on viruses is being reappraised. Safety in virology research is paramount and oversight frameworks should be reviewed periodically. Changes should be made with care, however, to avoid impeding science that is essential for rapidly reducing and responding to pandemic threats as well as addressing more common challenges caused by infectious diseases. Decades of research uniquely positioned the US to be able to respond to the COVID-19 crisis with astounding speed, delivering life-saving vaccines within a year of identifying the virus. We should embolden and empower this strength, which is a vital part of protecting the health, economy, and security of US citizens. Herein, we offer our perspectives on priorities for revised rules governing virology research in the US.


Subject(s)
Biomedical Research , Containment of Biohazards , Virology , Humans , COVID-19 , United States , Viruses , Biomedical Research/standards
9.
Proc Natl Acad Sci U S A ; 119(15): e2119531119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35394863

ABSTRACT

The RNA-binding protein RIG-I is a key initiator of the antiviral innate immune response. The signaling that mediates the antiviral response downstream of RIG-I is transduced through the adaptor protein MAVS and results in the induction of type I and III interferons (IFNs). This signal transduction occurs at endoplasmic reticulum (ER)­mitochondrial contact sites, to which RIG-I and other signaling proteins are recruited following their activation. RIG-I signaling is highly regulated to prevent aberrant activation of this pathway and dysregulated induction of IFN. Previously, we identified UFL1, the E3 ligase of the ubiquitin-like modifier conjugation system called ufmylation, as one of the proteins recruited to membranes at ER­mitochondrial contact sites in response to RIG-I activation. Here, we show that UFL1, as well as the process of ufmylation, promote IFN induction in response to RIG-I activation. We found that following RNA virus infection, UFL1 is recruited to the membrane-targeting protein 14­3-3ε and that this complex is then recruited to activated RIG-I to promote downstream innate immune signaling. Importantly, we found that 14­3-3ε has an increase in UFM1 conjugation following RIG-I activation. Additionally, loss of cellular ufmylation prevents the interaction of 14­3-3ε with RIG-I, which abrogates the interaction of RIG-I with MAVS and thus the downstream signal transduction that induces IFN. Our results define ufmylation as an integral regulatory component of the RIG-I signaling pathway and as a posttranslational control for IFN induction.


Subject(s)
DEAD Box Protein 58 , Interferons , RNA Virus Infections , RNA, Viral , Receptors, Immunologic , Ubiquitin-Protein Ligases , 14-3-3 Proteins/metabolism , DEAD Box Protein 58/metabolism , Humans , Immunity, Innate , Interferons/metabolism , RNA Virus Infections/genetics , RNA Virus Infections/immunology , RNA, Viral/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/metabolism
10.
Immunol Rev ; 304(1): 169-180, 2021 11.
Article in English | MEDLINE | ID: mdl-34405413

ABSTRACT

Induction of the antiviral innate immune response is highly regulated at the RNA level, particularly by RNA modifications. Recent discoveries have revealed how RNA modifications play key roles in cellular surveillance of nucleic acids and in controlling gene expression in response to viral infection. These modifications have emerged as being essential for a functional antiviral response and maintaining cellular homeostasis. In this review, we will highlight these and other discoveries that describe how the antiviral response is controlled by modifications to both viral and cellular RNA, focusing on how mRNA cap modifications, N6-methyladenosine, and RNA editing all contribute to coordinating an efficient response that properly controls viral infection.


Subject(s)
Immunity, Innate , Virus Diseases , Adenosine , Antiviral Agents/therapeutic use , Humans , RNA , RNA, Viral/genetics
11.
PLoS Biol ; 19(7): e3001342, 2021 07.
Article in English | MEDLINE | ID: mdl-34329302

ABSTRACT

A new study in PLOS Biology finds that interferon (IFN)-induced adenosine deaminase acting on RNA 1 (ADAR1) mRNA is N6-methyladenosine (m6A) modified to promote its translation, enabling ADAR1 to modify self-double-stranded RNAs (dsRNAs) generated during the IFN response and preventing activation of the melanoma differentiation-associated protein 5 (MDA5)-mediated host antiviral response.


Subject(s)
Adenosine Deaminase , RNA, Double-Stranded , Adenosine , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Interferons/metabolism , RNA, Double-Stranded/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
12.
J Virol ; 96(22): e0099722, 2022 11 23.
Article in English | MEDLINE | ID: mdl-36314819

ABSTRACT

Modification of the hepatitis C virus (HCV) positive-strand RNA genome by N6-methyladenosine (m6A) regulates the viral life cycle. This life cycle takes place solely in the cytoplasm, while m6A addition on cellular mRNA takes place in the nucleus. Thus, the mechanisms by which m6A is deposited on the viral RNA have been unclear. In this work, we find that m6A modification of HCV RNA by the m6A-methyltransferase proteins methyltransferase-like 3 and 14 (METTL3 and METTL14) is regulated by Wilms' tumor 1-associating protein (WTAP). WTAP, a predominantly nuclear protein, is an essential member of the cellular mRNA m6A-methyltransferase complex and known to target METTL3 to mRNA. We found that HCV infection induces localization of WTAP to the cytoplasm. Importantly, we found that WTAP is required for both METTL3 interaction with HCV RNA and m6A modification across the viral RNA genome. Further, we found that WTAP, like METTL3 and METTL14, negatively regulates the production of infectious HCV virions, a process that we have previously shown is regulated by m6A. Excitingly, WTAP regulation of both HCV RNA m6A modification and virion production was independent of its ability to localize to the nucleus. Together, these results reveal that WTAP is critical for HCV RNA m6A modification by METTL3 and METTL14 in the cytoplasm. IMPORTANCE Positive-strand RNA viruses such as HCV represent a significant global health burden. Previous work has described that HCV RNA contains the RNA modification m6A and how this modification regulates viral infection. Yet, how this modification is targeted to HCV RNA has remained unclear due to the incompatibility of the nuclear cellular processes that drive m6A modification with the cytoplasmic HCV life cycle. In this study, we present evidence for how m6A modification is targeted to HCV RNA in the cytoplasm by a mechanism in which WTAP recruits the m6A-methyltransferase METTL3 to HCV RNA. This targeting strategy for m6A modification of cytoplasmic RNA viruses is likely relevant for other m6A-modified positive-strand RNA viruses with cytoplasmic life cycles such as enterovirus 71 and SARS-CoV-2 and provides an exciting new target for potential antiviral therapies.


Subject(s)
Cell Cycle Proteins , Hepatitis C , Methyltransferases , RNA Splicing Factors , Humans , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C/genetics , Hepatitis C/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , RNA Splicing Factors/metabolism , RNA, Messenger/genetics , RNA, Viral/genetics , RNA, Viral/metabolism
13.
PLoS Pathog ; 15(2): e1007163, 2019 02.
Article in English | MEDLINE | ID: mdl-30730994

ABSTRACT

Hepatitis C virus (HCV) assembly and envelopment are coordinated by a complex protein interaction network that includes most of the viral structural and nonstructural proteins. While the nonstructural protein 4A (NS4A) is known to be important for viral particle production, the specific function of NS4A in this process is not well understood. We performed mutagenesis of the C-terminal acidic domain of NS4A and found that mutation of several of these amino acids prevented the formation of the viral envelope, and therefore the production of infectious virions, without affecting viral RNA replication. In an overexpression system, we found that NS4A interacted with several viral proteins known to coordinate envelopment, including the viral E1 glycoprotein. One of the NS4A C-terminal mutations, Y45F, disrupted the interaction of NS4A with E1. Specifically, NS4A interacted with the first hydrophobic region of E1, a region previously described as regulating viral particle production. Indeed, we found that an E1 mutation in this region, D72A, also disrupted the interaction of NS4A with E1. Supernatants from HCV NS4A Y45F transfected cells had significantly reduced levels of HCV RNA, however they contained equivalent levels of Core protein. Interestingly, the Core protein secreted from these cells formed high order oligomers with a density matching the infectious virus secreted from wild-type cells. These results suggest that this Y45F mutation in NS4A causes secretion of low-density Core particles lacking genomic HCV RNA. These results corroborate previous findings showing that the E1 D72A mutation also causes secretion of Core complexes lacking genomic HCV RNA, and therefore suggest that the interaction between NS4A and E1 is involved in the incorporation of viral RNA into infectious HCV particles. Our findings define a new role for NS4A in the HCV lifecycle and help elucidate the protein interactions necessary for production of infectious virus.


Subject(s)
Carrier Proteins/metabolism , Hepacivirus/physiology , Viral Envelope Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Base Sequence , Carrier Proteins/genetics , Cell Line , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C, Chronic/virology , Humans , Intracellular Signaling Peptides and Proteins , Mutation , Protein Domains , RNA, Viral , Viral Envelope Proteins/genetics , Viral Nonstructural Proteins/genetics , Virion/metabolism , Virion/physiology , Virus Assembly , Virus Replication
14.
Proc Natl Acad Sci U S A ; 115(35): 8829-8834, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30104368

ABSTRACT

N6-methyladenosine (m6A) RNA methylation is the most abundant epitranscriptomic modification of eukaryotic messenger RNAs (mRNAs). Previous reports have found m6A on both cellular and viral transcripts and defined its role in regulating numerous biological processes, including viral infection. Here, we show that m6A and its associated machinery regulate the life cycle of hepatitis B virus (HBV). HBV is a DNA virus that completes its life cycle via an RNA intermediate, termed pregenomic RNA (pgRNA). Silencing of enzymes that catalyze the addition of m6A to RNA resulted in increased HBV protein expression, but overall reduced reverse transcription of the pgRNA. We mapped the m6A site in the HBV RNA and found that a conserved m6A consensus motif situated within the epsilon stem loop structure, is the site for m6A modification. The epsilon stem loop is located in the 3' terminus of all HBV mRNAs and at both the 5' and 3' termini of the pgRNA. Mutational analysis of the identified m6A site in the 5' epsilon stem loop of pgRNA revealed that m6A at this site is required for efficient reverse transcription of pgRNA, while m6A methylation of the 3' epsilon stem loop results in destabilization of all HBV transcripts, suggesting that m6A has dual regulatory function for HBV RNA. Overall, this study reveals molecular insights into how m6A regulates HBV gene expression and reverse transcription, leading to an increased level of understanding of the HBV life cycle.


Subject(s)
Adenosine/analogs & derivatives , Gene Expression Regulation, Viral/physiology , Hepatitis B virus/physiology , Nucleic Acid Conformation , RNA Stability , RNA, Viral/biosynthesis , Adenosine/genetics , Adenosine/metabolism , Hep G2 Cells , Humans , RNA, Viral/genetics , Reverse Transcription/physiology , Viral Proteins/biosynthesis , Viral Proteins/genetics
15.
Proc Natl Acad Sci U S A ; 115(45): 11513-11518, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30341219

ABSTRACT

RNA virus genomes are efficient and compact carriers of biological information, encoding information required for replication both in their primary sequences and in higher-order RNA structures. However, the ubiquity of RNA elements with higher-order folds-in which helices pack together to form complex 3D structures-and the extent to which these elements affect viral fitness are largely unknown. Here we used single-molecule correlated chemical probing to define secondary and tertiary structures across the RNA genome of dengue virus serotype 2 (DENV2). Higher-order RNA structures are pervasive and involve more than one-third of nucleotides in the DENV2 genomic RNA. These 3D structures promote a compact overall architecture and contribute to viral fitness. Disrupting RNA regions with higher-order structures leads to stable, nonreverting mutants and could guide the development of vaccines based on attenuated RNA viruses. The existence of extensive regions of functional RNA elements with tertiary folds in viral RNAs, and likely many other messenger and noncoding RNAs, means that there are significant regions with pocket-containing surfaces that may serve as novel RNA-directed drug targets.


Subject(s)
Capsid/ultrastructure , Dengue Virus/ultrastructure , Genome, Viral , RNA, Viral/ultrastructure , Base Pairing , Capsid/chemistry , Capsid/metabolism , Dengue Virus/classification , Dengue Virus/genetics , Dengue Virus/metabolism , Genetic Fitness , Models, Molecular , Nucleic Acid Conformation , RNA, Viral/genetics , RNA, Viral/metabolism , Serogroup , Virus Assembly/genetics
16.
J Biol Chem ; 294(39): 14231-14240, 2019 09 27.
Article in English | MEDLINE | ID: mdl-31375559

ABSTRACT

Innate immune detection of viral nucleic acids during viral infection activates a signaling cascade that induces type I and type III IFNs as well as other cytokines, to generate an antiviral response. This signaling is initiated by pattern recognition receptors, such as the RNA helicase retinoic acid-inducible gene I (RIG-I), that sense viral RNA. These sensors then interact with the adaptor protein mitochondrial antiviral signaling protein (MAVS), which recruits additional signaling proteins, including TNF receptor-associated factor 3 (TRAF3) and TANK-binding kinase 1 (TBK1), to form a signaling complex that activates IFN regulatory factor 3 (IRF3) for transcriptional induction of type I IFNs. Here, using several immunological and biochemical approaches in multiple human cell types, we show that the GTPase-trafficking protein RAB1B up-regulates RIG-I pathway signaling and thereby promotes IFN-ß induction and the antiviral response. We observed that RAB1B overexpression increases RIG-I-mediated signaling to IFN-ß and that RAB1B deletion reduces signaling of this pathway. Additionally, loss of RAB1B dampened the antiviral response, indicated by enhanced Zika virus infection of cells depleted of RAB1B. Importantly, we identified the mechanism of RAB1B action in the antiviral response, finding that it forms a protein complex with TRAF3 to facilitate the interaction of TRAF3 with mitochondrial antiviral signaling protein. We conclude that RAB1B regulates TRAF3 and promotes the formation of innate immune signaling complexes in response to nucleic acid sensing during RNA virus infection.


Subject(s)
Immunity, Innate , TNF Receptor-Associated Factor 3/metabolism , Zika Virus Infection/immunology , rab1 GTP-Binding Proteins/metabolism , Animals , Chlorocebus aethiops , DEAD Box Protein 58/metabolism , HEK293 Cells , Humans , Interferon-beta/metabolism , Protein Binding , Receptors, Immunologic , Signal Transduction , Vero Cells
17.
J Virol ; 93(23)2019 12 01.
Article in English | MEDLINE | ID: mdl-31534039

ABSTRACT

The hepatitis C virus (HCV) NS3-NS4A protease complex is required for viral replication and is the major viral innate immune evasion factor. NS3-NS4A evades antiviral innate immunity by inactivating several proteins, including MAVS, the signaling adaptor for RIG-I and MDA5, and Riplet, an E3 ubiquitin ligase that activates RIG-I. Here, we identified a Tyr-16-Phe (Y16F) change in the NS4A transmembrane domain that prevents NS3-NS4A targeting of Riplet but not MAVS. This Y16F substitution reduces HCV replication in Huh7 cells, but not in Huh-7.5 cells, known to lack RIG-I signaling. Surprisingly, deletion of RIG-I in Huh7 cells did not restore Y16F viral replication. Rather, we found that Huh-7.5 cells lack Riplet expression and that the addition of Riplet to these cells reduced HCV Y16F replication, whereas the addition of Riplet lacking the RING domain restored HCV Y16F replication. In addition, TBK1 inhibition or IRF3 deletion in Huh7 cells was sufficient to restore HCV Y16F replication, and the Y16F protease lacked the ability to prevent IRF3 activation or interferon induction. Taken together, these data reveal that the NS4A Y16 residue regulates a noncanonical Riplet-TBK1-IRF3-dependent, but RIG-I-MAVS-independent, signaling pathway that limits HCV infection.IMPORTANCE The HCV NS3-NS4A protease complex facilitates viral replication by cleaving and inactivating the antiviral innate immune signaling proteins MAVS and Riplet, which are essential for RIG-I activation. NS3-NS4A therefore prevents IRF3 activation and interferon induction during HCV infection. Here, we uncover an amino acid residue within the NS4A transmembrane domain that is essential for inactivation of Riplet but does not affect MAVS cleavage by NS3-NS4A. Our study reveals that Riplet is involved in a RIG-I- and MAVS-independent signaling pathway that activates IRF3 and that this pathway is normally inactivated by NS3-NS4A during HCV infection. Our study selectively uncouples these distinct regulatory mechanisms within NS3-NS4A and defines a new role for Riplet in the antiviral response to HCV. Since Riplet is known to be inhibited by other RNA viruses, such as such influenza A virus, this innate immune signaling pathway may also be important in controlling other RNA virus infections.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepatitis C/virology , Serine Proteases/metabolism , Signal Transduction/drug effects , Viral Nonstructural Proteins/metabolism , Adaptor Proteins, Signal Transducing , Cell Line, Tumor , DEAD Box Protein 58/metabolism , Gene Knockout Techniques , HEK293 Cells , Hepatocytes/virology , Humans , Immune Evasion , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , Protein Serine-Threonine Kinases , Receptors, Immunologic , Virus Replication
19.
J Virol ; 89(14): 6974-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25948741

ABSTRACT

RNA virus infection is sensed in the cytoplasm by the retinoic acid-inducible gene I (RIG-I)-like receptors. These proteins signal through the host adaptor protein MAVS to trigger the antiviral innate immune response. Here, we describe how MAVS subcellular localization impacts its function and the regulation underlying MAVS signaling. We propose a model to describe how the coordination of MAVS functions at the interface between the mitochondria and the mitochondrion-associated endoplasmic reticulum (ER) membrane programs antiviral signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Immunity, Innate , RNA Viruses/immunology , Signal Transduction , Endoplasmic Reticulum/metabolism , Host-Pathogen Interactions , Mitochondria/metabolism , Models, Biological
20.
J Virol ; 89(22): 11523-33, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26355084

ABSTRACT

UNLABELLED: The molecular mechanisms that govern hepatitis C virus (HCV) assembly, release, and infectivity are still not yet fully understood. In the present study, we sequenced a genotype 2A strain of HCV (JFH-1) that had been cell culture adapted in Huh-7.5 cells to produce nearly 100-fold-higher viral titers than the parental strain. Sequence analysis identified nine mutations in the genome, present within both the structural and nonstructural genes. The infectious clone of this virus containing all nine culture-adapted mutations had 10-fold-higher levels of RNA replication and RNA release into the supernatant but had nearly 1,000-fold-higher viral titers, resulting in an increased specific infectivity compared to wild-type JFH-1. Two mutations, identified in the p7 polypeptide and NS5B RNA-dependent RNA polymerase, were sufficient to increase the specific infectivity of JFH-1. We found that the culture-adapted mutation in p7 promoted an increase in the size of cellular lipid droplets following transfection of viral RNA. In addition, we found that the culture-adaptive mutations in p7 and NS5B acted synergistically to enhance the specific viral infectivity of JFH-1 by decreasing the level of sphingomyelin in the virion. Overall, these results reveal a genetic interaction between p7 and NS5B that contributes to virion specific infectivity. Furthermore, our results demonstrate a novel role for the RNA-dependent RNA polymerase NS5B in HCV assembly. IMPORTANCE: Hepatitis C virus assembly and release depend on viral interactions with host lipid metabolic pathways. Here, we demonstrate that the viral p7 and NS5B proteins cooperate to promote virion infectivity by decreasing sphingomyelin content in the virion. Our data uncover a new role for the viral RNA-dependent RNA polymerase NS5B and p7 proteins in contributing to virion morphogenesis. Overall, these findings are significant because they reveal a genetic interaction between p7 and NS5B, as well as an interaction with sphingomyelin that regulates virion infectivity. Our data provide new strategies for targeting host lipid-virus interactions as potential targets for therapies against HCV infection.


Subject(s)
Hepacivirus/pathogenicity , Lipoproteins, HDL/metabolism , Sphingomyelins/metabolism , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism , Virion/pathogenicity , Base Sequence , Cell Line, Tumor , Hepacivirus/genetics , Hepacivirus/metabolism , Hepatitis C/virology , Humans , Lipid Droplets/physiology , RNA, Viral/biosynthesis , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Sequence Analysis, RNA , Viral Load/genetics , Viral Nonstructural Proteins/genetics , Viral Proteins/genetics , Virion/genetics , Virion/metabolism , Virus Assembly/genetics , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL