Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23.247
Filter
1.
PLoS Pathog ; 20(7): e1012345, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38968329

ABSTRACT

The CRISPR-Cas13 system has been proposed as an alternative treatment of viral infections. However, for this approach to be adopted as an antiviral, it must be optimized until levels of efficacy rival or exceed the performance of conventional approaches. To take steps toward this goal, we evaluated the influenza viral RNA degradation patterns resulting from the binding and enzymatic activity of mRNA-encoded LbuCas13a and two crRNAs from a prior study, targeting PB2 genomic and messenger RNA. We found that the genome targeting guide has the potential for significantly higher potency than originally detected, because degradation of the genomic RNA is not uniform across the PB2 segment, but it is augmented in proximity to the Cas13 binding site. The PB2 genome targeting guide exhibited high levels (>1 log) of RNA degradation when delivered 24 hours post-infection in vitro and maintained that level of degradation over time, with increasing multiplicity of infection (MOI), and across modern influenza H1N1 and H3N2 strains. Chemical modifications to guides with potent LbuCas13a function, resulted in nebulizer delivered efficacy (>1-2 log reduction in viral titer) in a hamster model of influenza (Influenza A/H1N1/California/04/09) infection given prophylactically or as a treatment (post-infection). Maximum efficacy was achieved with two doses, when administered both pre- and post-infection. This work provides evidence that mRNA-encoded Cas13a can effectively mitigate Influenza A infections opening the door to the development of a programmable approach to treating multiple respiratory infections.


Subject(s)
CRISPR-Cas Systems , Influenza, Human , RNA Stability , RNA, Messenger , RNA, Viral , Animals , RNA, Viral/genetics , RNA, Viral/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Humans , Influenza, Human/virology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Orthomyxoviridae Infections/virology , Antiviral Agents/pharmacology , Dogs , Cricetinae , Viral Proteins/genetics , Viral Proteins/metabolism , Mesocricetus , Madin Darby Canine Kidney Cells
2.
Nature ; 631(8021): 670-677, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987591

ABSTRACT

In all organisms, regulation of gene expression must be adjusted to meet cellular requirements and frequently involves helix-turn-helix (HTH) domain proteins1. For instance, in the arms race between bacteria and bacteriophages, rapid expression of phage anti-CRISPR (acr) genes upon infection enables evasion from CRISPR-Cas defence; transcription is then repressed by an HTH-domain-containing anti-CRISPR-associated (Aca) protein, probably to reduce fitness costs from excessive expression2-5. However, how a single HTH regulator adjusts anti-CRISPR production to cope with increasing phage genome copies and accumulating acr mRNA is unknown. Here we show that the HTH domain of the regulator Aca2, in addition to repressing Acr synthesis transcriptionally through DNA binding, inhibits translation of mRNAs by binding conserved RNA stem-loops and blocking ribosome access. The cryo-electron microscopy structure of the approximately 40 kDa Aca2-RNA complex demonstrates how the versatile HTH domain specifically discriminates RNA from DNA binding sites. These combined regulatory modes are widespread in the Aca2 family and facilitate CRISPR-Cas inhibition in the face of rapid phage DNA replication without toxic acr overexpression. Given the ubiquity of HTH-domain-containing proteins, it is anticipated that many more of them elicit regulatory control by dual DNA and RNA binding.


Subject(s)
Bacteriophages , CRISPR-Cas Systems , Cryoelectron Microscopy , Models, Molecular , Bacteriophages/metabolism , Bacteriophages/genetics , Bacteriophages/chemistry , CRISPR-Cas Systems/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/chemistry , Protein Biosynthesis , Helix-Turn-Helix Motifs , Ribosomes/metabolism , Ribosomes/chemistry , Binding Sites , Protein Domains , Viral Proteins/metabolism , Viral Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Associated Proteins/chemistry , Nucleic Acid Conformation , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA, Viral/metabolism , RNA, Viral/genetics , RNA, Viral/chemistry , Transcription, Genetic
3.
PLoS Biol ; 22(7): e3002709, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39012844

ABSTRACT

RNA viruses have notoriously high mutation rates due to error-prone replication by their RNA polymerase. However, natural selection concentrates variability in a few key viral proteins. To test whether this stems from different mutation tolerance profiles among viral proteins, we measured the effect of >40,000 non-synonymous mutations across the full proteome of coxsackievirus B3 as well as >97% of all possible codon deletions in the nonstructural proteins. We find significant variation in mutational tolerance within and between individual viral proteins, which correlated with both general and protein-specific structural and functional attributes. Furthermore, mutational fitness effects remained stable across cell lines, suggesting selection pressures are mostly conserved across environments. In addition to providing a rich dataset for understanding virus biology and evolution, our results illustrate that incorporation of mutational tolerance data into druggable pocket discovery can aid in selecting targets with high barriers to drug resistance.


Subject(s)
Enterovirus B, Human , Mutation , Proteome , Enterovirus B, Human/genetics , Proteome/metabolism , Humans , Viral Proteins/genetics , Viral Proteins/metabolism , Genetic Fitness , Virus Replication/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 121(30): e2403805121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39018195

ABSTRACT

It is commonly held that there is a fundamental relationship between genome size and error rate, manifest as a notional "error threshold" that sets an upper limit on genome sizes. The genome sizes of RNA viruses, which have intrinsically high mutation rates due to a lack of mechanisms for error correction, must therefore be small to avoid accumulating an excessive number of deleterious mutations that will ultimately lead to population extinction. The proposed exceptions to this evolutionary rule are RNA viruses from the order Nidovirales (such as coronaviruses) that encode error-correcting exonucleases, enabling them to reach genome lengths greater than 40 kb. The recent discovery of large-genome flavi-like viruses (Flaviviridae), which comprise genomes up to 27 kb in length yet seemingly do not encode exonuclease domains, has led to the proposal that a proofreading mechanism is required to facilitate the expansion of nonsegmented RNA virus genomes above 30 kb. Herein, we describe a ~40 kb flavi-like virus identified in a Haliclona sponge metatranscriptome that does not encode a known exonuclease. Structural analysis revealed that this virus may have instead captured cellular domains associated with nucleic acid metabolism that have not been previously found in RNA viruses. Phylogenetic inference placed this virus as a divergent pesti-like lineage, such that we have provisionally termed it "Maximus pesti-like virus." This virus represents an instance of a flavi-like virus achieving a genome size comparable to that of the Nidovirales and demonstrates that RNA viruses have evolved multiple solutions to overcome the error threshold.


Subject(s)
Genome, Viral , Animals , Phylogeny , Genome Size , Viral Proteins/genetics , Viral Proteins/metabolism , Exonucleases/metabolism , Exonucleases/genetics , RNA, Viral/genetics
5.
Front Immunol ; 15: 1363156, 2024.
Article in English | MEDLINE | ID: mdl-38953028

ABSTRACT

Introduction: Human Herpesvirus 6B (HHV-6B) impedes host immune responses by downregulating class I MHC molecules (MHC-I), hindering antigen presentation to CD8+ T cells. Downregulation of MHC-I disengages inhibitory receptors on natural killer (NK) cells, resulting in activation and killing of the target cell if NK cell activating receptors such as NKG2D have engaged stress ligands upregulated on the target cells. Previous work has shown that HHV-6B downregulates three MHC-like stress ligands MICB, ULBP1, and ULBP3, which are recognized by NKG2D. The U20 glycoprotein of the related virus HHV-6A has been implicated in the downregulation of ULBP1, but the precise mechanism remains undetermined. Methods: We set out to investigate the role of HHV-6B U20 in modulating NK cell activity. We used HHV-6B U20 expressed as a recombinant protein or transduced into target cells, as well as HHV-6B infection, to investigate binding interactions with NK cell ligands and receptors and to assess effects on NK cell activation. Small-angle X-ray scattering was used to align molecular models derived from machine-learning approaches. Results: We demonstrate that U20 binds directly to ULBP1 with sub-micromolar affinity. Transduction of U20 decreases NKG2D binding to ULBP1 at the cell surface but does not decrease ULBP1 protein levels, either at the cell surface or in toto. HHV-6B infection and soluble U20 have the same effect. Transduction of U20 blocks NK cell activation in response to cell-surface ULBP1. Structural modeling of the U20 - ULBP1 complex indicates some similarities to the m152-RAE1γ complex.


Subject(s)
GPI-Linked Proteins , Herpesvirus 6, Human , Killer Cells, Natural , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Herpesvirus 6, Human/immunology , GPI-Linked Proteins/metabolism , GPI-Linked Proteins/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , NK Cell Lectin-Like Receptor Subfamily K/immunology , Lymphocyte Activation/immunology , Protein Binding , Viral Proteins/immunology , Viral Proteins/metabolism , Glycoproteins/immunology , Glycoproteins/metabolism , Intracellular Signaling Peptides and Proteins
6.
Subcell Biochem ; 104: 181-205, 2024.
Article in English | MEDLINE | ID: mdl-38963488

ABSTRACT

Tailed double-stranded DNA bacteriophage employs a protein terminase motor to package their genome into a preformed protein shell-a system shared with eukaryotic dsDNA viruses such as herpesviruses. DNA packaging motor proteins represent excellent targets for antiviral therapy, with Letermovir, which binds Cytomegalovirus terminase, already licensed as an effective prophylaxis. In the realm of bacterial viruses, these DNA packaging motors comprise three protein constituents: the portal protein, small terminase and large terminase. The portal protein guards the passage of DNA into the preformed protein shell and acts as a protein interaction hub throughout viral assembly. Small terminase recognises the viral DNA and recruits large terminase, which in turn pumps DNA in an ATP-dependent manner. Large terminase also cleaves DNA at the termination of packaging. Multiple high-resolution structures of each component have been resolved for different phages, but it is only more recently that the field has moved towards cryo-EM reconstructions of protein complexes. In conjunction with highly informative single-particle studies of packaging kinetics, these structures have begun to inspire models for the packaging process and its place among other DNA machines.


Subject(s)
DNA, Viral , Viral Proteins , DNA, Viral/genetics , DNA, Viral/metabolism , Viral Proteins/metabolism , Viral Proteins/genetics , Endodeoxyribonucleases/metabolism , Endodeoxyribonucleases/genetics , Viral Genome Packaging/physiology , DNA Packaging , Bacteriophages/genetics , Bacteriophages/physiology , Bacteriophages/metabolism , Genome, Viral
7.
J Gen Virol ; 105(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38959049

ABSTRACT

Phasmaviridae is a family for negative-sense RNA viruses with genomes of about 9.7-15.8 kb. These viruses are maintained in and/or transmitted by insects. Phasmavirids produce enveloped virions containing three single-stranded RNA segments that encode a nucleoprotein (N), a glycoprotein precursor (GPC), and a large (L) protein containing an RNA-directed RNA polymerase (RdRP) domain. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Phasmaviridae, which is available at ictv.global/report/phasmaviridae.


Subject(s)
Genome, Viral , RNA, Viral , Animals , RNA, Viral/genetics , Negative-Sense RNA Viruses/genetics , Negative-Sense RNA Viruses/classification , Virion/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Insecta/virology , Phylogeny , Virus Replication
8.
Vet Res ; 55(1): 86, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970119

ABSTRACT

H7N9 subtype avian influenza viruses (AIVs) cause 1567 human infections and have high mortality, posing a significant threat to public health. Previously, we reported that two avian-derived H7N9 isolates (A/chicken/Eastern China/JTC4/2013 and A/chicken/Eastern China/JTC11/2013) exhibit different pathogenicities in mice. To understand the genetic basis for the differences in virulence, we constructed a series of mutant viruses based on reverse genetics. We found that the PB2-E627K mutation alone was not sufficient to increase the virulence of H7N9 in mice, despite its ability to enhance polymerase activity in mammalian cells. However, combinations with PB1-V719M and/or PA-N444D mutations significantly enhanced H7N9 virulence. Additionally, these combined mutations augmented polymerase activity, thereby intensifying virus replication, inflammatory cytokine expression, and lung injury, ultimately increasing pathogenicity in mice. Overall, this study revealed that virulence in H7N9 is a polygenic trait and identified novel virulence-related residues (PB2-627K combined with PB1-719M and/or PA-444D) in viral ribonucleoprotein (vRNP) complexes. These findings provide new insights into the molecular mechanisms underlying AIV pathogenesis in mammals, with implications for pandemic preparedness and intervention strategies.


Subject(s)
Influenza A Virus, H7N9 Subtype , Mutation , Orthomyxoviridae Infections , Viral Proteins , Animals , Mice , Influenza A Virus, H7N9 Subtype/genetics , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza A Virus, H7N9 Subtype/physiology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/veterinary , Virulence , Female , Viral Proteins/genetics , Viral Proteins/metabolism , Mice, Inbred BALB C , Virus Replication
9.
Front Cell Infect Microbiol ; 14: 1418168, 2024.
Article in English | MEDLINE | ID: mdl-38988816

ABSTRACT

Exosomes are extracelluar vesicles that facilitate intercellular communication and are pivotal in post-transcriptional regulation within cellular gene regulatory networks, impacting pathogen dynamics. These vesicles serve as crucial regulators of immune responses, mediating cellular interactions and enabling the introduction of viral pathogenic regions into host cells. Exosomes released from virus-infected cells harbor diverse microRNAs (miRNAs), which can be transferred to recipient cells, thereby modulating virus infection. This transfer is a critical element in the molecular interplay mediated by exosomes. Additionally, the endosomal sorting complex required for transport (ESCRT) within exosomes plays a vital role in virus infection, with ESCRT components binding to viral proteins to facilitate virus budding. This review elucidates the roles of exosomes and their constituents in the invasion of host cells by viruses, aiming to shed new light on the regulation of viral transmission via exosomes.


Subject(s)
Endosomal Sorting Complexes Required for Transport , Exosomes , Host-Pathogen Interactions , MicroRNAs , Virus Diseases , Exosomes/metabolism , Humans , Endosomal Sorting Complexes Required for Transport/metabolism , Virus Diseases/metabolism , Virus Diseases/virology , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Viruses/pathogenicity , Viruses/metabolism , Virus Release , Viral Proteins/metabolism , Viral Proteins/genetics
10.
Cell Host Microbe ; 32(7): 1039-1041, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38991498

ABSTRACT

Bacteria have evolved anti-viral defenses, but the mechanisms of sensing and stopping infection are still under investigation. In this issue of Cell Host & Microbe, Mets, Kurata, Ernits et al. describe how direct sensing of a phage protein by a bacterial toxin-antitoxin-associated chaperone unleashes toxin activity to prevent infection.


Subject(s)
Bacteriophages , Molecular Chaperones , Molecular Chaperones/metabolism , Bacteriophages/physiology , Toxin-Antitoxin Systems , Bacterial Toxins/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/genetics , Bacteria/virology , Bacteria/metabolism , Bacteria/genetics
11.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39000573

ABSTRACT

Mycobacteriophages are viruses that specifically infect bacterial species within the genera Mycobacterium and Mycolicibacterium. Over 2400 mycobacteriophages have been isolated on the host Mycolicibacterium smegmatis and sequenced. This wealth of genomic data indicates that mycobacteriophage genomes are diverse, mosaic, and contain numerous (35-60%) genes for which there is no predicted function based on sequence similarity to characterized orthologs, many of which are essential to lytic growth. To fully understand the molecular aspects of mycobacteriophage-host interactions, it is paramount to investigate the function of these genes and gene products. Here we show that the temperate mycobacteriophage, Alexphander, makes stable lysogens with a frequency of 2.8%. Alexphander gene 94 is essential for lytic infection and encodes a protein predicted to contain a C-terminal MerR family helix-turn-helix DNA-binding motif (HTH) and an N-terminal DinB/YfiT motif, a putative metal-binding motif found in stress-inducible gene products. Full-length and C-terminal gp94 constructs form high-order nucleoprotein complexes on 100-500 base pair double-stranded DNA fragments and full-length phage genomic DNA with little sequence discrimination for the DNA fragments tested. Maximum gene 94 mRNA levels are observed late in the lytic growth cycle, and gene 94 is transcribed in a message with neighboring genes 92 through 96. We hypothesize that gp94 is an essential DNA-binding protein for Alexphander during lytic growth. We proposed that gp94 forms multiprotein complexes on DNA through cooperative interactions involving its HTH DNA-binding motif at sites throughout the phage chromosome, facilitating essential DNA transactions required for lytic propagation.


Subject(s)
DNA-Binding Proteins , Mycobacteriophages , Mycobacterium smegmatis , Viral Proteins , Mycobacteriophages/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mycobacterium smegmatis/virology , Mycobacterium smegmatis/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/chemistry , Lysogeny/genetics , Genome, Viral , DNA, Viral/genetics
12.
Science ; 385(6704): 105-112, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38963841

ABSTRACT

Introns containing homing endonucleases are widespread in nature and have long been assumed to be selfish elements that provide no benefit to the host organism. These genetic elements are common in viruses, but whether they confer a selective advantage is unclear. In this work, we studied intron-encoded homing endonuclease gp210 in bacteriophage ΦPA3 and found that it contributes to viral competition by interfering with the replication of a coinfecting phage, ΦKZ. We show that gp210 targets a specific sequence in ΦKZ, which prevents the assembly of progeny viruses. This work demonstrates how a homing endonuclease can be deployed in interference competition among viruses and provide a relative fitness advantage. Given the ubiquity of homing endonucleases, this selective advantage likely has widespread evolutionary implications in diverse plasmid and viral competition as well as virus-host interactions.


Subject(s)
Endonucleases , Introns , Pseudomonas Phages , Pseudomonas aeruginosa , Viral Interference , Viral Proteins , Endonucleases/metabolism , Endonucleases/genetics , Viral Interference/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Assembly , Virus Replication , Pseudomonas Phages/enzymology , Pseudomonas Phages/genetics , Pseudomonas aeruginosa/virology
13.
J Biomed Sci ; 31(1): 70, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003473

ABSTRACT

Coronaviruses employ various strategies for survival, among which the activation of endogenous or exogenous apoptosis stands out, with viral proteins playing a pivotal role. Notably, highly pathogenic coronaviruses such as SARS-CoV-2, SARS-CoV, and MERS-CoV exhibit a greater array of non-structural proteins compared to low-pathogenic strains, facilitating their ability to induce apoptosis via multiple pathways. Moreover, these viral proteins are adept at dampening host immune responses, thereby bolstering viral replication and persistence. This review delves into the intricate interplay between highly pathogenic coronaviruses and apoptosis, systematically elucidating the molecular mechanisms underpinning apoptosis induction by viral proteins. Furthermore, it explores the potential therapeutic avenues stemming from apoptosis inhibition as antiviral agents and the utilization of apoptosis-inducing viral proteins as therapeutic modalities. These insights not only shed light on viral pathogenesis but also offer novel perspectives for cancer therapy.


Subject(s)
Apoptosis , SARS-CoV-2 , Humans , SARS-CoV-2/physiology , Viral Proteins/metabolism , Viral Proteins/genetics , Middle East Respiratory Syndrome Coronavirus/physiology , Severe acute respiratory syndrome-related coronavirus/physiology , COVID-19/virology
14.
J Med Virol ; 96(7): e29752, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38949191

ABSTRACT

Antiviral signaling, immune response and cell metabolism are dysregulated by SARS-CoV-2, the causative agent of COVID-19. Here, we show that SARS-CoV-2 accessory proteins ORF3a, ORF9b, ORF9c and ORF10 induce a significant mitochondrial and metabolic reprogramming in A549 lung epithelial cells. While ORF9b, ORF9c and ORF10 induced largely overlapping transcriptomes, ORF3a induced a distinct transcriptome, including the downregulation of numerous genes with critical roles in mitochondrial function and morphology. On the other hand, all four ORFs altered mitochondrial dynamics and function, but only ORF3a and ORF9c induced a marked alteration in mitochondrial cristae structure. Genome-Scale Metabolic Models identified both metabolic flux reprogramming features both shared across all accessory proteins and specific for each accessory protein. Notably, a downregulated amino acid metabolism was observed in ORF9b, ORF9c and ORF10, while an upregulated lipid metabolism was distinctly induced by ORF3a. These findings reveal metabolic dependencies and vulnerabilities prompted by SARS-CoV-2 accessory proteins that may be exploited to identify new targets for intervention.


Subject(s)
COVID-19 , Mitochondria , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , Mitochondria/metabolism , COVID-19/metabolism , COVID-19/virology , COVID-19/pathology , A549 Cells , Viral Regulatory and Accessory Proteins/metabolism , Viral Regulatory and Accessory Proteins/genetics , Transcriptome , Open Reading Frames , Viral Proteins/genetics , Viral Proteins/metabolism , Viroporin Proteins
15.
Microb Biotechnol ; 17(7): e14513, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38962879

ABSTRACT

The phage lysin field has done nothing but grow in the last decades. As a result, many different research groups around the world are contributing to the field, often with certain methodological differences that pose a challenge to the interpretation and comparison of results. In this work, we present the case study of three Acinetobacter baumannii-targeting phage lysins (wild-type endolysin LysMK34 plus engineered lysins eLysMK34 and 1D10) plus one lysin with broad activity against Gram-positive bacteria (PlySs2) to provide exemplary evidence on the risks of generalization when using one of the most common lysin evaluation assays: the killing assay with resting cells. To that end, we performed killing assays with the aforementioned lysins using hypo-, iso- and hypertonic buffers plus human serum either as the reaction or the dilution medium in a systematic manner. Our findings stress the perils of creating hypotonic conditions or a hypotonic shock during a killing assay, suggesting that hypotonic buffers should be avoided as a test environment or as diluents before plating to avoid overestimation of the killing effect in the assayed conditions. As a conclusion, we suggest that the nature of both the incubation and the dilution buffers should be always clearly identified when reporting killing activity data, and that for experimental consistency the same incubation buffer should be used as a diluent for posterior serial dilution and plating unless explicitly required by the experimental design. In addition, the most appropriate buffer mimicking the final application must be chosen to obtain relevant results.


Subject(s)
Acinetobacter baumannii , Bacteriophages , Bacteriophages/chemistry , Bacteriophages/physiology , Bacteriophages/genetics , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/virology , Osmolar Concentration , Microbial Viability/drug effects , Buffers , Humans , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Proteins/chemistry , Endopeptidases/metabolism , Endopeptidases/chemistry
16.
BMC Microbiol ; 24(1): 267, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030475

ABSTRACT

BACKGROUND: Grapevine fanleaf virus (GFLV) is one of the most detrimental viral pathogens of grapevines worldwide but no information is available on its effect on the root system architecture (RSA) of plant hosts. We used two wildtype GFLV strains and their single amino acid mutants to assess RSA traits in infected Nicotiana benthamiana and evaluate transcriptomic changes in host root gene expression in replicated time course 3'RNA-Seq experiments. Mutations targeted the multi-functional GFLV-encoded protein 1EPol*/Sd, a putative RNA-dependent RNA polymerase and determinant of foliar symptoms in N. benthamiana plants. RESULTS: Plant infection with wildtype GFLV strain GHu and mutant GFLV strain F13 1EPol G802K, both carrying a lysine in position 802 of protein 1EPol*/Sd, resulted in a significantly lower number of root tips (-30%), and a significantly increased average root diameter (+ 20%) at 17 days post inoculation (dpi) in comparison with roots of mock inoculated plants. In contrast, the RSA of plants infected with wildtype GFLV strain F13 and mutant GFLV strain GHu 1EPol K802G, both carrying a glycine in position 802 of protein 1EPol*/Sd, resembled that of mock inoculated plants. Modifications of RSA traits were not associated with GFLV titer. Root tissue transcriptome analysis at 17 dpi indicated dysregulation of pattern recognition receptors, plant hormones, RNA silencing, and genes related to the production of reactive oxygen species (ROS). For wildtype GFLV strain GHu, RSA modifications were correlated with an abundant accumulation of ROS in the pericycle of primary roots at 7 dpi and the duration of vein clearing symptom expression in apical leaves. Dysegulation of a hypersensitive response was an overarching gene ontology found through enrichment analyses of 3'RNA-Seq data. CONCLUSIONS: Our findings revealed the causative role of lysine in position 802 of protein 1EPol*/Sd in a novel RSA phenotype during viral infection and documented GFLV-N. benthamiana interactions at the root level based on (i) antiviral response, (ii) receptor mediated production of ROS, and (iii) hormone regulation. A correlation between above and below ground symptoms was reported for the first time in plants infected with wildtype GFLV strain GHu. Further work is warranted to test whether the modified RSA of a plant host might impact GFLV acquisition and transmission by the ectoparasitic dagger nematode Xiphinema index.


Subject(s)
Nicotiana , Plant Diseases , Plant Roots , Plant Roots/virology , Plant Roots/genetics , Plant Diseases/virology , Plant Diseases/genetics , Nicotiana/virology , Nicotiana/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Nepovirus/genetics , Host-Pathogen Interactions , Mutation , Gene Expression Regulation, Plant , Vitis/virology , Vitis/genetics , Amino Acids/metabolism , Plant Leaves/virology , Plant Leaves/genetics , Transcriptome
17.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000098

ABSTRACT

Potato mop-top virus (PMTV) is an emerging viral pathogen that causes tuber necrosis in potatoes. PMTV is composed of three single-stranded RNA segments: RNA1 encodes RNA-dependent RNA polymerase, RNA2 contains the coat protein (CP), and RNA3 harbors a triple gene block (TGB 1, TGB2, and TGB3). CP plays a role in viral transmission, while TGB is known to facilitate cell-to-cell and long-distance systemic movement. The role of CP in symptom development, specifically in the presence of TGB genes, was investigated using potato virus X (PVX) as a delivery vehicle to express PMTV genes in the model plant Nicotiana benthamiana. Plants expressing individual genes showed mild symptoms that included leaf curling and crumpling. Interestingly, symptom severity varied among plants infected with three different combinations: CP with TGB1, CP with TGB2, and CP with TGB3. Notably, the combination of CP and TGB3 induced a hypersensitive response, accompanied by stunted growth and downward curling and crumpling. These results suggest the potential role of TGB co-expressed with CP in symptom development during PMTV infection. Additionally, this study demonstrates the use of the PVX-based expression system as a valuable platform for assessing the role of unknown genes in viral pathogenicity.


Subject(s)
Capsid Proteins , Nicotiana , Plant Diseases , Potexvirus , Solanum tuberosum , Capsid Proteins/genetics , Capsid Proteins/metabolism , Nicotiana/genetics , Nicotiana/virology , Nicotiana/metabolism , Potexvirus/genetics , Potexvirus/pathogenicity , Plant Diseases/virology , Plant Diseases/genetics , Solanum tuberosum/virology , Solanum tuberosum/genetics , Solanum tuberosum/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
18.
Molecules ; 29(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38893337

ABSTRACT

mRNA vaccines are entering a period of rapid development. However, their synthesis is still plagued by challenges related to mRNA impurities and fragments (incomplete mRNA). Most impurities of mRNA products transcribed in vitro are mRNA fragments. Only full-length mRNA transcripts containing both a 5'-cap and a 3'-poly(A) structure are viable for in vivo expression. Therefore, RNA fragments are the primary product-related impurities that significantly hinder mRNA efficacy and must be effectively controlled; these species are believed to originate from either mRNA hydrolysis or premature transcriptional termination. In the manufacturing of commercial mRNA vaccines, T7 RNA polymerase-catalyzed in vitro transcription (IVT) synthesis is a well-established method for synthesizing long RNA transcripts. This study identified a pivotal domain on the T7 RNA polymerase that is associated with erroneous mRNA release. By leveraging the advantageous properties of a T7 RNA polymerase mutant and precisely optimized IVT process parameters, we successfully achieved an mRNA integrity exceeding 91%, thereby further unlocking the immense potential of mRNA therapeutics.


Subject(s)
DNA-Directed RNA Polymerases , RNA, Messenger , Transcription, Genetic , Viral Proteins , RNA, Messenger/genetics , DNA-Directed RNA Polymerases/metabolism , DNA-Directed RNA Polymerases/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , mRNA Vaccines
19.
Emerg Microbes Infect ; 13(1): 2369193, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38873898

ABSTRACT

The global outbreak of Mpox, caused by the monkeypox virus (MPXV), has attracted international attention and become another major infectious disease event after COVID-19. The mRNA cap N7 methyltransferase (RNMT) of MPXV methylates the N7 position of the added guanosine to the 5'-cap structure of mRNAs and plays a vital role in evading host antiviral immunity. MPXV RNMT is composed of the large subunit E1 and the small subunit E12. How E1 and E12 of MPXV assembly remains unclear. Here, we report the crystal structures of E12, the MTase domain of E1 with E12 (E1CTD-E12) complex, and the E1CTD-E12-SAM ternary complex, revealing the detailed conformations of critical residues and the structural changes upon E12 binding to E1. Functional studies suggest that E1CTD N-terminal extension (Asp545-Arg562) and the small subunit E12 play an essential role in the binding process of SAM. Structural comparison of the AlphaFold2-predicted E1, E1CTD-E12 complex, and the homologous D1-D12 complex of vaccinia virus (VACV) indicates an allosteric activating effect of E1 in MPXV. Our findings provide the structural basis for the MTase activity stimulation of the E1-E12 complex and suggest a potential interface for screening the anti-poxvirus inhibitors.


Subject(s)
Methyltransferases , Monkeypox virus , Methyltransferases/chemistry , Methyltransferases/metabolism , Methyltransferases/genetics , Monkeypox virus/genetics , Monkeypox virus/enzymology , Monkeypox virus/chemistry , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Crystallography, X-Ray , RNA Caps/metabolism , RNA Caps/chemistry , Models, Molecular , Humans , Protein Conformation , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/chemistry
20.
PLoS Pathog ; 20(6): e1012311, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38885273

ABSTRACT

The majority of rod-shaped and some filamentous plant viruses encode a cysteine-rich protein (CRP) that functions in viral virulence; however, the roles of these CRPs in viral infection remain largely unknown. Here, we used barley stripe mosaic virus (BSMV) as a model to investigate the essential role of its CRP in virus morphogenesis. The CRP protein γb directly interacts with BSMV coat protein (CP), the mutations either on the His-85 site in γb predicted to generate a potential CCCH motif or on the His-13 site in CP exposed to the surface of the virions abolish the zinc-binding activity and their interaction. Immunogold-labeling assays show that γb binds to the surface of rod-shaped BSMV virions in a Zn2+-dependent manner, which enhances the RNA binding activity of CP and facilitates virion assembly and stability, suggesting that the Zn2+-dependent physical association of γb with the virion is crucial for BSMV morphogenesis. Intriguingly, the tightly binding of diverse CRPs to their rod-shaped virions is a general feature employed by the members in the families Virgaviridae (excluding the genus Tobamovirus) and Benyviridae. Together, these results reveal a hitherto unknown role of CRPs in the assembly and stability of virus particles, and expand our understanding of the molecular mechanism underlying virus morphogenesis.


Subject(s)
Virion , Zinc , Zinc/metabolism , Virion/metabolism , Capsid Proteins/metabolism , Virus Assembly/physiology , Plant Viruses/metabolism , Plant Viruses/physiology , Plant Diseases/virology , Cysteine/metabolism , Viral Proteins/metabolism , Morphogenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...