Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.550
Filter
1.
Nat Commun ; 15(1): 8528, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39358425

ABSTRACT

Usutu virus (USUV) and West Nile virus (WNV) are two closely related emerging mosquito-borne flaviviruses. Their natural hosts are wild birds, but they can also cause severe neurological disorders in humans. Both viruses are efficiently suppressed by type I interferon (IFN), which interferes with viral replication, dissemination, pathogenesis and transmission. Here, we show that the replication of USUV and WNV are inhibited through a common set of IFN-induced genes (ISGs), with the notable exception of ISG20, which USUV is resistant to. Strikingly, USUV was the only virus among all the other tested mosquito-borne flaviviruses that demonstrated resistance to the 3'-5' exonuclease activity of ISG20. Our findings highlight that the intrinsic resistance of the USUV genome, irrespective of the presence of cellular or viral proteins or protective post-transcriptional modifications, relies on a unique sequence present in its 3' untranslated region. Importantly, this genomic region alone can confer ISG20 resistance to a susceptible flavivirus, without compromising its infectivity, suggesting that it could be acquired by other flaviviruses. This study provides new insights into the strategy employed by emerging flaviviruses to overcome host defense mechanisms.


Subject(s)
3' Untranslated Regions , Flavivirus , Virus Replication , West Nile virus , 3' Untranslated Regions/genetics , Flavivirus/genetics , Flavivirus/physiology , Humans , Animals , Virus Replication/genetics , West Nile virus/genetics , West Nile virus/physiology , Flavivirus Infections/virology , Exonucleases/metabolism , Exonucleases/genetics , Chlorocebus aethiops , Exoribonucleases/metabolism , Exoribonucleases/genetics , HEK293 Cells , Vero Cells , Cell Line , Interferon Type I/metabolism , Genome, Viral
2.
Methods Enzymol ; 705: 81-109, 2024.
Article in English | MEDLINE | ID: mdl-39389674

ABSTRACT

There is growing recognition that viral RNA genomes possess enzymatically incorporated modified nucleosides. These small chemical changes are analogous to epigenomic modifications in DNA and have the potential to be similarly important modulators of viral transcription and evolution. However, the molecular level consequences of individual sites of modification remain to be broadly explored. Here we describe an in vitro assay to examine the impact of nucleoside modifications on the rate and fidelity of SARS-CoV-2 RNA transcription. Establishing the role of modified nucleotides in SARS-CoV-2 is of interest both for advancing fundamental knowledge of RNA modifications in viruses, and because modulating the modification-landscape of SARS-CoV-2 may represent a therapeutic strategy to interfere with viral RNA replication. Our approach can be used to assess the influence both of modifications present in a template RNA, as well nucleotide analog inhibitors. These methods provide a reproducible guide for generating active SARS-CoV-2 replication/transcription complexes capable of establishing how RNA modifications influence the pre-steady state rate constants of nucleotide addition by RNA-dependent RNA polymerases.


Subject(s)
Nucleosides , RNA, Viral , SARS-CoV-2 , Virus Replication , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Nucleosides/metabolism , Nucleosides/chemistry , Humans , Virus Replication/genetics , Viral Transcription/genetics , COVID-19/virology , COVID-19/metabolism , Transcription, Genetic
3.
Nat Commun ; 15(1): 8017, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39271696

ABSTRACT

The HIV-1 capsid is composed of capsid (CA) protein hexamers and pentamers (capsomers) that contain a central pore hypothesised to regulate capsid assembly and facilitate nucleotide import early during post-infection. These pore functions are mediated by two positively charged rings created by CA Arg-18 (R18) and Lys-25 (K25). Here we describe the forced evolution of viruses containing mutations in R18 and K25. Whilst R18 mutants fail to replicate, K25A viruses acquire compensating mutations that restore nearly wild-type replication fitness. These compensating mutations, which rescue reverse transcription and infection without reintroducing lost pore charges, map to three adaptation hot-spots located within and between capsomers. The second-site suppressor mutations act by restoring the formation of pentamers lost upon K25 mutation, enabling closed conical capsid assembly both in vitro and inside virions. These results indicate that there is no intrinsic requirement for K25 in either nucleotide import or capsid assembly. We propose that whilst HIV-1 must maintain a precise hexamer:pentamer equilibrium for proper capsid assembly, compensatory mutations can tune this equilibrium to restore fitness lost by mutation of the central pore.


Subject(s)
Capsid Proteins , Capsid , HIV-1 , Mutation , Virus Assembly , Virus Replication , HIV-1/genetics , HIV-1/physiology , Virus Assembly/genetics , Capsid Proteins/genetics , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Capsid/metabolism , Humans , Virus Replication/genetics , Virion/metabolism , Virion/genetics , HEK293 Cells , HIV Infections/virology , HIV Infections/genetics
4.
Mem Inst Oswaldo Cruz ; 119: e240071, 2024.
Article in English | MEDLINE | ID: mdl-39292108

ABSTRACT

BACKGROUND: Human immunodeficiency virus (HIV)-1 infection can activate the expression of human endogenous retroviruses (HERVs), particularly HERV-K (HML-2). HIV controllers (HICs) are rare people living with HIV (PLWHs) who naturally control HIV-1 replication and overexpress some cellular restriction factors that negatively regulate the LTR-driven transcription of HIV-1 proviruses. OBJECTIVES: To understand the ability of HICs to control the expression of endogenous retroviruses. METHODS: We measured endogenous retrovirus type K6 (ERVK-6) RNA expression in peripheral blood mononuclear cells (PBMCs) of HICs (n = 23), antiretroviral (ART)-suppressed subjects (n = 8), and HIV-1-negative (NEG) individuals (n = 10) and correlated the transcript expression of ERVK-6 with multiple HIV-1 cellular restriction factors. FINDINGS: Our study revealed that ERVK-6 RNA expression in PBMCs from HICs was significantly downregulated compared with that in both the ART and NEG control groups. Moreover, we detected that ERVK-6 RNA levels in PBMCs across all groups were negatively correlated with the expression levels of p21 and MCPIP1, two cellular restriction factors that limit the activation of macrophages and T cells by downregulating the activity of NF-kB. MAIN CONCLUSIONS: These findings support the hypothesis that HICs activate innate antiviral mechanisms that may simultaneously downregulate the transcription of both exogenous (HIV-1) and endogenous (ERVK-6) retroviruses. Future studies with larger cohorts should be performed to confirm this hypothesis and to explore the role of p21 and MCPIP1 in regulating HERV-K expression in physiological and pathological conditions.


Subject(s)
Endogenous Retroviruses , HIV Infections , HIV-1 , RNA, Viral , Ribonucleases , Adult , Female , Humans , Male , Middle Aged , Case-Control Studies , Cyclin-Dependent Kinase Inhibitor p21/genetics , Endogenous Retroviruses/genetics , Endogenous Retroviruses/immunology , HIV Infections/immunology , HIV Infections/virology , HIV Infections/genetics , HIV-1/genetics , HIV-1/immunology , Immunity, Innate/genetics , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Ribonucleases/genetics , Ribonucleases/metabolism , RNA, Viral/genetics , Transcription Factors/genetics , Virus Replication/genetics
5.
Proc Natl Acad Sci U S A ; 121(40): e2407990121, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39320912

ABSTRACT

We explored how a simple retrovirus, Mason-Pfizer monkey virus (M-PMV) to facilitate its replication process, utilizes DHX15, a cellular RNA helicase, typically engaged in RNA processing. Through advanced genetic engineering techniques, we showed that M-PMV recruits DHX15 by mimicking cellular mechanisms, relocating it from the nucleus to the cytoplasm to aid in viral assembly. This interaction is essential for the correct packaging of the viral genome and critical for its infectivity. Our findings offer unique insights into the mechanisms of viral manipulation of host cellular processes, highlighting a sophisticated strategy that viruses employ to leverage cellular machinery for their replication. This study adds valuable knowledge to the understanding of viral-host interactions but also suggests a common evolutionary history between cellular processes and viral mechanisms. This finding opens a unique perspective on the export mechanism of intron-retaining mRNAs in the packaging of viral genetic information and potentially develop ways to stop it.


Subject(s)
Mason-Pfizer monkey virus , RNA, Viral , Virus Assembly , Animals , Humans , Cell Nucleus/metabolism , Cell Nucleus/virology , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/genetics , Genome, Viral , HEK293 Cells , Mason-Pfizer monkey virus/genetics , Mason-Pfizer monkey virus/metabolism , Mason-Pfizer monkey virus/physiology , RNA Helicases/metabolism , RNA Helicases/genetics , RNA, Viral/metabolism , RNA, Viral/genetics , Virus Assembly/genetics , Virus Assembly/physiology , Virus Replication/genetics , Virus Replication/physiology
6.
Proc Natl Acad Sci U S A ; 121(39): e2408078121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39292744

ABSTRACT

The Pentamer complex of Human Cytomegalovirus (HCMV) consists of the viral glycoproteins gH, gL, UL128, UL130, and UL131 and is incorporated into infectious virions. HCMV strains propagated extensively in vitro in fibroblasts carry UL128, UL130, or UL131 alleles that do not make a functional complex and thus lack Pentamer function. Adding functional Pentamer to such strains decreases virus growth in fibroblasts. Here, we show that the Pentamer inhibits productive HCMV replication in fibroblasts by repressing viral Immediate Early (IE) transcription. We show that ectopic expression of the viral IE1 protein, a target of Pentamer-mediated transcriptional repression, complements the growth defect of a Pentamer-positive virus. Furthermore, we show that the Pentamer also represses viral IE transcription in cell types where HCMV in vitro latency is studied. Finally, we identify UL130 as a functional subunit of the Pentamer for IE transcriptional repression and demonstrate that cyclic AMP Response Element (CRE) and NFkB sites within the Major Immediate Early Promoter that drives IE1 transcription contribute to this repression. We conclude that the HCMV Pentamer represses viral IE transcription.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Fibroblasts , Immediate-Early Proteins , Transcription, Genetic , Viral Envelope Proteins , Humans , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Cytomegalovirus/metabolism , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/metabolism , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/genetics , Fibroblasts/virology , Fibroblasts/metabolism , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Gene Expression Regulation, Viral , Virus Replication/genetics , Glycoproteins/metabolism , Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Genes, Immediate-Early , Promoter Regions, Genetic
7.
Int J Mol Sci ; 25(18)2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39337625

ABSTRACT

MicroRNAs (miRNAs) are molecules that influence messenger RNA (mRNA) expression levels by binding to the 3' untranslated region (3' UTR) of target genes. Host miRNAs can influence flavivirus replication, either by inducing changes in the host transcriptome or by directly binding to viral genomes. The 3' UTR of the flavivirus genome is a conserved region crucial for viral replication. Cells might exploit this well-preserved region by generating miRNAs that interact with it, ultimately impacting viral replication. Despite significant efforts to identify miRNAs capable of arresting viral replication, the potential of all these miRNAs to interact with the flavivirus 3' UTR is still poorly characterised. In this context, bioinformatic tools have been proposed as a fundamental part of accelerating the discovery of interactions between miRNAs and the 3' UTR of viral genomes. In this study, we performed a computational analysis to reveal potential miRNAs from human and mosquito species that bind to the 3' UTR of flaviviruses. In humans, miR-6842 and miR-661 were found, while in mosquitoes, miR-9-C, miR-2945-5p, miR-11924, miR-282-5p, and miR-79 were identified. These findings open new avenues for studying these miRNAs as antivirals against flavivirus infections.


Subject(s)
3' Untranslated Regions , Computational Biology , Flavivirus , Genome, Viral , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Flavivirus/genetics , Humans , Animals , Computational Biology/methods , Virus Replication/genetics , Antiviral Agents/pharmacology , Flavivirus Infections/virology , Flavivirus Infections/genetics , Culicidae/virology , Culicidae/genetics
8.
Nat Commun ; 15(1): 7640, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223139

ABSTRACT

Genetic parasites, including viruses and transposons, exploit components from the host for their own replication. However, little is known about virus-transposon interactions within host cells. Here, we discover a strategy where human cytomegalovirus (HCMV) hijacks L1 retrotransposon encoded protein during its replication cycle. HCMV infection upregulates L1 expression by enhancing both the expression of L1-activating transcription factors, YY1 and RUNX3, and the chromatin accessibility of L1 promoter regions. Increased L1 expression, in turn, promotes HCMV replicative fitness. Affinity proteomics reveals UL44, HCMV DNA polymerase subunit, as the most abundant viral binding protein of the L1 ribonucleoprotein (RNP) complex. UL44 directly interacts with L1 ORF2p, inducing DNA damage responses in replicating HCMV compartments. While increased L1-induced mutagenesis is not observed in HCMV for genetic adaptation, the interplay between UL44 and ORF2p accelerates viral DNA replication by alleviating replication stress. Our findings shed light on how HCMV exploits host retrotransposons for enhanced viral fitness.


Subject(s)
Cytomegalovirus , DNA Replication , Long Interspersed Nucleotide Elements , Viral Proteins , Virus Replication , Humans , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Virus Replication/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , DNA Replication/genetics , Long Interspersed Nucleotide Elements/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/genetics , Host-Pathogen Interactions/genetics , Retroelements/genetics , DNA-Binding Proteins
9.
Signal Transduct Target Ther ; 9(1): 243, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39289355

ABSTRACT

The various mutations in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pose a substantial challenge in mitigating the viral infectivity. The identification of novel host factors influencing SARS-CoV-2 replication holds potential for discovering new targets for broad-spectrum antiviral drugs that can combat future viral mutations. In this study, potential host factors regulated by SARS-CoV-2 infection were screened through different high-throughput sequencing techniques and further identified in cells. Subsequent analysis and experiments showed that the reduction of m6A modification level on ACTN4 (Alpha-actinin-4) mRNA leads to a decrease in mRNA stability and translation efficiency, ultimately inhibiting ACTN4 expression. In addition, ACTN4 was demonstrated to target nsp12 for binding and characterized as a competitor for SARS-CoV-2 RNA and the RNA-dependent RNA polymerase complex, thereby impeding viral replication. Furthermore, two ACTN4 agonists, YS-49 and demethyl-coclaurine, were found to dose-dependently inhibit SARS-CoV-2 infection in both Huh7 cells and K18-hACE2 transgenic mice. Collectively, this study unveils the pivotal role of ACTN4 in SARS-CoV-2 infection, offering novel insights into the intricate interplay between the virus and host cells, and reveals two potential candidates for future anti-SARS-CoV-2 drug development.


Subject(s)
Actinin , Antiviral Agents , COVID-19 Drug Treatment , SARS-CoV-2 , Virus Replication , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , Humans , Animals , Antiviral Agents/pharmacology , Actinin/genetics , Actinin/metabolism , Mice , Virus Replication/drug effects , Virus Replication/genetics , COVID-19/virology , COVID-19/genetics , COVID-19/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Mice, Transgenic , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Coronavirus RNA-Dependent RNA Polymerase/genetics , RNA, Viral/genetics
10.
Int J Mol Sci ; 25(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39125583

ABSTRACT

Coronaviruses constitute a global threat to human and animal health. It is essential to investigate the long-distance RNA-RNA interactions that approximate remote regulatory elements in strategies, including genome circularization, discontinuous transcription, and transcriptional enhancers, aimed at the rapid replication of their large genomes, pathogenicity, and immune evasion. Based on the primary sequences and modeled RNA-RNA interactions of two experimentally defined coronaviral enhancers, we detected via an in silico primary and secondary structural analysis potential enhancers in various coronaviruses, from the phylogenetically ancient avian infectious bronchitis virus (IBV) to the recently emerged SARS-CoV-2. These potential enhancers possess a core duplex-forming region that could transition between closed and open states, as molecular switches directed by viral or host factors. The duplex open state would pair with remote sequences in the viral genome and modulate the expression of downstream crucial genes involved in viral replication and host immune evasion. Consistently, variations in the predicted IBV enhancer region or its distant targets coincide with cases of viral attenuation, possibly driven by decreased open reading frame (ORF)3a immune evasion protein expression. If validated experimentally, the annotated enhancer sequences could inform structural prediction tools and antiviral interventions.


Subject(s)
Enhancer Elements, Genetic , Genome, Viral , Infectious bronchitis virus , SARS-CoV-2 , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Infectious bronchitis virus/genetics , Humans , Enhancer Elements, Genetic/genetics , Animals , RNA, Viral/genetics , RNA, Viral/metabolism , COVID-19/virology , COVID-19/genetics , Betacoronavirus/genetics , Virus Replication/genetics , Coronavirus Infections/virology , Transcription, Genetic , Gene Expression Regulation, Viral , Pneumonia, Viral/virology
11.
Nucleic Acids Res ; 52(17): 10654-10667, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39087525

ABSTRACT

Chikungunya virus (CHIKV) is a re-emerging, pathogenic alphavirus that is transmitted to humans by Aedesspp. mosquitoes-causing fever and debilitating joint pain, with frequent long-term health implications and high morbidity. The CHIKV replication cycle is poorly understood and specific antiviral therapeutics are lacking. In the current study, we identify host cell Musashi RNA binding protein-2 (MSI-2) as a proviral factor. MSI-2 depletion and small molecule inhibition assays demonstrated that MSI-2 is required for efficient CHIKV genome replication. Depletion of both MSI-2 and MSI-1 homologues was found to synergistically inhibit CHIKV replication, suggesting redundancy in their proviral function. Electromobility shift assay (EMSA) competition studies demonstrated that MSI-2 interacts specifically with an RNA binding motif within the 5' untranslated region (5'UTR) of CHIKV and reverse genetic analysis showed that mutation of the binding motif inhibited genome replication and blocked rescue of mutant virus. For the first time, this study identifies the proviral role of MSI RNA binding proteins in the replication of the CHIKV genome, providing important new insight into mechanisms controlling replication of this significant human pathogen and the potential of a novel therapeutic target.


Subject(s)
5' Untranslated Regions , Chikungunya virus , Genome, Viral , RNA-Binding Proteins , Virus Replication , Chikungunya virus/genetics , Virus Replication/genetics , Humans , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , 5' Untranslated Regions/genetics , Animals , RNA, Viral/metabolism , RNA, Viral/genetics , Chikungunya Fever/virology , Chikungunya Fever/genetics , HEK293 Cells , Protein Binding , Cell Line
12.
J Mol Biol ; 436(20): 168738, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39117177

ABSTRACT

The mouse mammary tumor virus (MMTV) is a well-known causative agent of breast cancer in mice. Previously, we have shown that MMTV dysregulates expression of the host miR-17-92 cluster in MMTV-infected mammary glands and MMTV-induced tumors. This cluster, better known as oncomiR-1, is frequently dysregulated in cancers, particularly breast cancer. In this study, our aim was to uncover a functional interaction between MMTV and the cluster. Our results reveal that MMTV expression led to dysregulation of the cluster in both mammary epithelial HC11 and HEK293T cells with the expression of miR-92a cluster member being affected the most. Conversely, overexpression of the whole or partial cluster significantly repressed MMTV expression. Notably, overexpression of cluster member miR-92a alone repressed MMTV expression to the same extent as overexpression of the complete/partial cluster. Inhibition of miR-92a led to nearly a complete restoration of MMTV expression, while deletion/substitution of the miR-92a seed sequence rescued MMTV expression. Dual luciferase assays identified MMTV genomic RNA as the potential target of miR-92a. These results show that the miR-17-92 cluster acts as part of the cell's well-known miRNA-based anti-viral response to thwart incoming MMTV infection. Thus, this study provides the first evidence highlighting the biological significance of host miRNAs in regulating MMTV replication and potentially influencing tumorigenesis.


Subject(s)
Mammary Tumor Virus, Mouse , MicroRNAs , Virus Replication , MicroRNAs/genetics , MicroRNAs/metabolism , Mammary Tumor Virus, Mouse/genetics , Virus Replication/genetics , Animals , Humans , Mice , HEK293 Cells , Multigene Family , Host-Pathogen Interactions/genetics , Female , Cell Line
13.
Microbiol Spectr ; 12(9): e0071424, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39109857

ABSTRACT

The emergence and spread of the African swine fever virus (ASFV) posed a significant threat to the global swine breeding industry, calling for innovative approaches benefiting viral containment and control. A recent study (Z. Zheng, L. Xu, H. Dou, Y. Zhou, X., et al., Microbiol Spectr 12: e02164-23, 2024, https://doi.org/10.1128/spectrum.02164-23) established a multiplexed CRISPR-Cas system targeting the genome of ASFV and tested the consequent antiviral activity both in vitro and in vivo. Application of this system showed a significant reduction of viral replication in vitro, while the germline-edited pigs expressing this system exhibited normal growth with continuous guide RNA expression. Although no survival advantage was observed upon ASFV challenge compared with nonengineered pigs, this marks the first attempt of germline editing to pursue ASFV resistance and paves the way for future disease-resistant animal breeding approaches utilizing CRISPR-Cas technology.


Subject(s)
African Swine Fever Virus , African Swine Fever , CRISPR-Cas Systems , Gene Editing , Animals , African Swine Fever Virus/genetics , Swine , African Swine Fever/virology , Gene Editing/methods , Virus Replication/genetics , Genome, Viral/genetics , Disease Resistance/genetics
14.
Nat Commun ; 15(1): 7602, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39217162

ABSTRACT

Rift Valley fever virus (RVFV) is a mosquito-borne zoonotic pathogen. Its RNA genome consists of two negative-sense segments (L and M) with one gene each, and one ambisense segment (S) with two opposing genes separated by the noncoding "intergenic region" (IGR). These vRNAs and the complementary cRNAs are encapsidated by nucleoprotein (N). Using iCLIP2 (individual-nucleotide resolution UV crosslinking and immunoprecipitation) to map all N-vRNA and N-cRNA interactions, we detect N coverage along the L and M segments. However, the S segment vRNA and cRNA each contain approximately 100 non-encapsidated nucleotides stretching from the IGR into the 5'-adjacent reading frame. These exposed regions are RNase-sensitive and predicted to form stem-loop structures with the mRNA transcription termination motif positioned near the top. Moreover, optimal S segment transcription and replication requires the entire exposed region rather than only the IGR. Thus, the RVFV S segment contains a central, non-encapsidated RNA region with a functional role.


Subject(s)
RNA, Viral , Rift Valley fever virus , Rift Valley fever virus/genetics , RNA, Viral/genetics , Animals , DNA, Intergenic/genetics , Genome, Viral , Virus Replication/genetics , Rift Valley Fever/virology , Rift Valley Fever/transmission , Nucleic Acid Conformation , Nucleoproteins/genetics , Nucleoproteins/metabolism , Humans , Transcription, Genetic
15.
EMBO Rep ; 25(9): 4078-4090, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39122863

ABSTRACT

The human immunodeficiency virus (HIV-1) is highly dependent on a variety of host factors. Beside proteins, host RNA molecules are reported to aid HIV-1 replication and latency maintenance. Here, we implement multiple workflows of native RNA immunoprecipitation and sequencing (nRIPseq) to determine direct host RNA interaction partners of all 18 HIV-1 (poly)proteins. We identify 1,727 HIV-1 protein - human RNA interactions in the Jurkat cell line and 1,558 interactions in SupT1 cells for a subset of proteins, and discover distinct cellular pathways that seem to be used or controlled by HIV-1 on the RNA level: Tat binds mRNAs of proteins involved in the super elongation complex (AFF1-4, Cyclin-T1). Correlation of the interaction scores (based on binding abundancy) allows identifying the highest confidence interactions, for which we perform a small-scale knockdown screen that leads to the identification of three HIV-1 protein binding RNA interactors involved in HIV-1 replication (AFF2, H4C9 and RPLP0).


Subject(s)
HIV-1 , Proteome , Virus Replication , Humans , HIV-1/genetics , HIV-1/metabolism , HIV-1/physiology , Jurkat Cells , Proteome/metabolism , Virus Replication/genetics , Host-Pathogen Interactions/genetics , Protein Binding , Cyclin T/metabolism , Cyclin T/genetics , RNA, Messenger/metabolism , RNA, Messenger/genetics , HIV Infections/virology , HIV Infections/metabolism , HIV Infections/genetics , RNA, Viral/metabolism , RNA, Viral/genetics , tat Gene Products, Human Immunodeficiency Virus/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomal Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics
16.
Genes (Basel) ; 15(8)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39202352

ABSTRACT

Atypical porcine pestivirus (APPV) can cause congenital tremor type A-II in neonatal piglets, posing a significant threat to swine herd health globally. Our previous study demonstrated that the Mut domains, comprising 112 amino acids at the N-terminus, are the primary functional regions of the E2 protein of APPV. This study identified 14 host cellular proteins that exhibit potential interactions with the Mut domains of the E2 protein using yeast two-hybrid screening. Using bioinformatics analysis, we discovered that the Mut domains of the E2 protein might exert regulatory effects on apoptosis by modulating energy metabolism within the mitochondria. We also conducted co-immunoprecipitation, glutathione S-transferase pull-down, and immunofluorescence assays to confirm the interaction between the Mut domains of the E2 protein and cathepsin H and signal sequence receptor subunit 4 (SSR4). Ultimately, SSR4 enhanced APPV replication in vitro. In summary, our study successfully elucidated the interactions between the Mut domains of the E2 protein and host cell protein, predicted the potential pathways implicated in these interactions, and demonstrated SSR4 involvement in APPV infection. These significant findings contribute valuable knowledge toward a deeper understanding of APPV pathogenesis and the role of the Mut domains of the E2 protein in this intricate process.


Subject(s)
Pestivirus Infections , Pestivirus , Animals , Pestivirus/genetics , Pestivirus/metabolism , Swine , Pestivirus Infections/veterinary , Pestivirus Infections/virology , Pestivirus Infections/genetics , Swine Diseases/virology , Swine Diseases/genetics , Swine Diseases/metabolism , Host-Pathogen Interactions/genetics , Protein Domains , Virus Replication/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Humans , Protein Interaction Maps/genetics
17.
Microbiol Spectr ; 12(10): e0082924, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39166862

ABSTRACT

The heterogeneous nuclear ribonucleoprotein (hnRNP A2B1) is a key component of the hnRNP complex involving RNA modulation in eukaryotic cells and it has also been reported to be involved in the replication of the hepatitis E virus, influenza A virus, and hepatitis B virus. However, it is not clear whether the role of the hnRNP A2B1 in viral replication is conserved among RNA viruses and what is the mechanism of hnRNP A2B1 in RNA virus replication. In this study, we first used severe fever with thrombocytopenia syndrome virus (SFTSV), a tick-borne RNA virus that causes a severe viral hemorrhagic fever as well as other RNA viruses including VSV-GFP, SeV, EV71, and ZIKV to demonstrate that knockout hnRNPA2B1 gene inhibited viral RNA replication and overexpression of hnRNP A2B1 could restore the RNA levels of all tested RNA viruses. These results suggest that hnRNPA2B1 upregulation of viral replication is conserved among RNA viruses. Next, we demonstrated that hnRNP A2B1 was translocated from the nucleus to the cytoplasm under RNA virus infection including SFTSV, VSV-GFP, SeV, EV71, and ZIKV, suggesting translocation of hnRNP A2B1 from the nucleus to the cytoplasm is crucial for RNA virus replication. We then used SFTSV as a model to demonstrate the mechanism of hnRNP A2B1 in the promotion of RNA virus replication. We found that overexpression of SFTSV nucleoprotein can also cause hnRNP A2B1 translocation from the nucleus to the cytoplasm and that the SFTSV NP interacted with the RNA recognition motif 1 domain of hnRNP A2B1. We further demonstrated that the hnRNP A2B1 interacted with the 5' UTR of SFTSV RNA. In conclusion, we revealed that the hnRNP A2B1 upregulation of viral RNA replication is conserved among RNA viruses; the mechanism of hnRNP A2B1 in promotion of SFTSV viral RNA replication is that SFTSV NP interacted with the hnRNPA2B1 to retain it in the cytoplasm where the hnRNP A2B1 interacted with the 5' UTR of SFTSV RNA to promote the viral RNA replication.IMPORTANCESevere fever with thrombocytopenia syndrome virus (SFTSV) is a tick-borne RNA virus with a high mortality rate of up to 30%. In this study, we first used SFTSV as a model to demonstrate that the role of hnRNPA2B1 in viral replication is conserved in SFTSV. Then we used other RNA viruses, including VSV-GFP, SeV, EV71, and ZIKV, to repeat the experiment and demonstrated the same results as SFTSV in all tested RNA viruses. By knocking out the hnRNPA2B1 gene, SFTSV RNA replication was inhibited, and overexpression of hnRNPA2B1 restored RNA levels of SFTSV and other tested RNA viruses. We revealed a novel mechanism where the SFTSV nucleoprotein interacts with hnRNPA2B1, retaining it in the cytoplasm. This interaction promotes viral RNA replication by binding to the 5' UTR of SFTSV RNA. The findings suggest that targeting hnRNPA2B1 could be a potential strategy for developing broad-spectrum antiviral therapies, given its conserved role across different RNA viruses. This research provides significant insights into the replication mechanisms of RNA viruses and highlights potential targets for antiviral interventions.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Phlebovirus , RNA Viruses , RNA, Viral , Virus Replication , Animals , Humans , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/virology , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Phlebovirus/genetics , Phlebovirus/physiology , RNA Viruses/genetics , RNA Viruses/physiology , RNA, Viral/genetics , RNA, Viral/metabolism , Severe Fever with Thrombocytopenia Syndrome/virology , Severe Fever with Thrombocytopenia Syndrome/genetics , Severe Fever with Thrombocytopenia Syndrome/metabolism , Virus Replication/genetics , Mice
18.
Nucleic Acids Res ; 52(17): 10311-10328, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39126317

ABSTRACT

A major function of the DNA damage responses (DDRs) that act during the replicative phase of the cell cycle is to inhibit initiation and elongation of DNA replication. It has been shown that DNA replication of the polyomavirus, SV40, is inhibited and its replication fork is slowed by cellular DDR responses. The inhibition of SV40 DNA replication is associated with enhanced DDR kinase phosphorylation of SV40 Large T-antigen (LT), the viral DNA helicase. Mass spectroscopy was used to identify a novel highly conserved DDR kinase site, T518, on LT. In cell-based assays expression of a phosphomimetic form of LT at T518 (T518D) resulted in dramatically decreased levels of SV40 DNA replication, but LT-dependent transcriptional activation was unaffected. Purified WT and LT T518D were analyzed in vitro. In concordance with the cell-based data, reactions using SV40 LT-T518D, but not T518A, showed dramatic inhibition of SV40 DNA replication. A myriad of LT protein-protein interactions and LT's biochemical functions were unaffected by the LT T518D mutation; however, LT's DNA helicase activity was dramatically decreased on long, but not very short, DNA templates. These results suggest that DDR phosphorylation at T518 inhibits SV40 DNA replication by suppressing LT helicase activity.


Subject(s)
DNA Damage , DNA Helicases , DNA Replication , Simian virus 40 , Phosphorylation , Simian virus 40/genetics , Humans , DNA Helicases/metabolism , DNA Helicases/genetics , Antigens, Polyomavirus Transforming/metabolism , Antigens, Polyomavirus Transforming/genetics , Virus Replication/genetics , Cell Line
19.
Nat Commun ; 15(1): 7353, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39191758

ABSTRACT

G-quadruplex (G4) structures are found in eukaryotic cell replication origins, but their role in origin function remains unclear. In this study G4 motifs are found in the lytic DNA replication origin (oriLyt) of human cytomegalovirus (HCMV) and recombinant viruses show that a G4 motif in oriLyt essential region I (ER-I) is necessary for viral growth. Replication assays of oriLyt-containing plasmids and biochemical/biophysical analyses show that G4 formation in ER-I is crucial for viral DNA replication. G4 pull-down analysis identifies viral DNA replication factors, such as IE2, UL84, and UL44, as G4-binding proteins. In enzyme-linked immunosorbent assays, specific G4-binding ligands inhibit G4 binding by the viral proteins. The Epstein-Barr virus oriLyt core element also forms a stable G4 that could substitute for the oriLyt ER-I G4 in HCMV. These results demonstrate that viral G4s in replication origins represent an essential structural element in recruiting replication factors and might be a therapeutic target against viral infections.


Subject(s)
Cytomegalovirus , DNA Replication , DNA, Viral , G-Quadruplexes , Immediate-Early Proteins , Replication Origin , Viral Proteins , Virus Replication , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Humans , Virus Replication/genetics , Replication Origin/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Immediate-Early Proteins/metabolism , Immediate-Early Proteins/genetics , DNA, Viral/genetics , DNA, Viral/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Protein Binding
20.
Sci Rep ; 14(1): 19594, 2024 08 23.
Article in English | MEDLINE | ID: mdl-39179600

ABSTRACT

Coronavirus (CoV) possesses numerous functional cis-acting elements in its positive-strand genomic RNA. Although most of these RNA structures participate in viral replication, the functions of RNA structures in the genomic RNA of CoV in viral replication remain unclear. In this study, we investigated the functions of the higher-order RNA stem-loop (SL) structures SL5B, SL5C, and SL5D in the ORF1a coding region of Middle East respiratory syndrome coronavirus (MERS-CoV) in viral replication. Our approach, using reverse genetics of a bacterial artificial chromosome system, revealed that SL5B and SL5C play essential roles in the discontinuous transcription of MERS-CoV. In silico analyses predicted that SL5C interacts with a bulged stem-loop (BSL) in the 3' untranslated region, suggesting that the RNA structure of SL5C is important for viral RNA transcription. Conversely, SL5D did not affect transcription, but mediated the synthesis of positive-strand genomic RNA. Additionally, the RNA secondary structure of SL5 in the revertant virus of the SL5D mutant was similar to that of the wild-type, indicating that the RNA structure of SL5D can finely tune RNA replication in MERS-CoV. Our data indicate novel regulatory mechanisms of viral RNA transcription and replication by higher-order RNA structures in the MERS-CoV genomic RNA.


Subject(s)
Middle East Respiratory Syndrome Coronavirus , Nucleic Acid Conformation , RNA, Viral , Transcription, Genetic , Virus Replication , Middle East Respiratory Syndrome Coronavirus/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Replication/genetics , Open Reading Frames/genetics , Humans , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , 3' Untranslated Regions/genetics , Animals
SELECTION OF CITATIONS
SEARCH DETAIL