Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23.264
Filtrer
1.
Microb Biotechnol ; 17(7): e14513, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38962879

RÉSUMÉ

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.


Sujet(s)
Acinetobacter baumannii , Bactériophages , Bactériophages/composition chimique , Bactériophages/physiologie , Bactériophages/génétique , Acinetobacter baumannii/effets des médicaments et des substances chimiques , Acinetobacter baumannii/virologie , Concentration osmolaire , Viabilité microbienne/effets des médicaments et des substances chimiques , Substances tampon , Humains , Protéines virales/génétique , Protéines virales/métabolisme , Protéines virales/composition chimique , Endopeptidases/métabolisme , Endopeptidases/composition chimique
2.
J Gen Virol ; 105(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38959049

RÉSUMÉ

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.


Sujet(s)
Génome viral , ARN viral , Animaux , ARN viral/génétique , Virus à ARN de polarité négative/génétique , Virus à ARN de polarité négative/classification , Virion/génétique , Protéines virales/génétique , Protéines virales/métabolisme , Insectes/virologie , Phylogenèse , Réplication virale
3.
Front Immunol ; 15: 1363156, 2024.
Article de Anglais | MEDLINE | ID: mdl-38953028

RÉSUMÉ

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.


Sujet(s)
Protéines liées au GPI , Herpèsvirus humain de type 6 , Cellules tueuses naturelles , Activation des lymphocytes , Sous-famille K des récepteurs de cellules NK de type lectine , Humains , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Herpèsvirus humain de type 6/immunologie , Protéines liées au GPI/métabolisme , Protéines liées au GPI/immunologie , Sous-famille K des récepteurs de cellules NK de type lectine/métabolisme , Sous-famille K des récepteurs de cellules NK de type lectine/immunologie , Activation des lymphocytes/immunologie , Liaison aux protéines , Protéines virales/immunologie , Protéines virales/métabolisme , Glycoprotéines/immunologie , Glycoprotéines/métabolisme , Protéines et peptides de signalisation intracellulaire
4.
Subcell Biochem ; 104: 181-205, 2024.
Article de Anglais | MEDLINE | ID: mdl-38963488

RÉSUMÉ

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.


Sujet(s)
ADN viral , Protéines virales , ADN viral/génétique , ADN viral/métabolisme , Protéines virales/métabolisme , Protéines virales/génétique , Endodeoxyribonucleases/métabolisme , Endodeoxyribonucleases/génétique , Encapsidation du génome viral/physiologie , Empaquetage de l'ADN , Bactériophages/génétique , Bactériophages/physiologie , Bactériophages/métabolisme , Génome viral
5.
Cell Host Microbe ; 32(7): 1039-1041, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38991498

RÉSUMÉ

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.


Sujet(s)
Bactériophages , Chaperons moléculaires , Chaperons moléculaires/métabolisme , Bactériophages/physiologie , Systèmes toxine-antitoxine , Toxines bactériennes/métabolisme , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Protéines virales/métabolisme , Protéines virales/génétique , Bactéries/virologie , Bactéries/métabolisme , Bactéries/génétique
6.
Proc Natl Acad Sci U S A ; 121(30): e2403805121, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39018195

RÉSUMÉ

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.


Sujet(s)
Génome viral , Animaux , Phylogenèse , Taille du génome , Protéines virales/génétique , Protéines virales/métabolisme , Exonucleases/métabolisme , Exonucleases/génétique , ARN viral/génétique
7.
PLoS Biol ; 22(7): e3002709, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39012844

RÉSUMÉ

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.


Sujet(s)
Entérovirus humain B , Mutation , Protéome , Entérovirus humain B/génétique , Protéome/métabolisme , Humains , Protéines virales/génétique , Protéines virales/métabolisme , Aptitude génétique , Réplication virale/génétique , Protéines virales non structurales/génétique , Protéines virales non structurales/métabolisme
8.
Vet Res ; 55(1): 86, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970119

RÉSUMÉ

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.


Sujet(s)
Sous-type H7N9 du virus de la grippe A , Mutation , Infections à Orthomyxoviridae , Protéines virales , Animaux , Souris , Sous-type H7N9 du virus de la grippe A/génétique , Sous-type H7N9 du virus de la grippe A/pathogénicité , Sous-type H7N9 du virus de la grippe A/physiologie , Infections à Orthomyxoviridae/virologie , Infections à Orthomyxoviridae/médecine vétérinaire , Virulence , Femelle , Protéines virales/génétique , Protéines virales/métabolisme , Souris de lignée BALB C , Réplication virale
9.
J Biomed Sci ; 31(1): 70, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39003473

RÉSUMÉ

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.


Sujet(s)
Apoptose , SARS-CoV-2 , Humains , SARS-CoV-2/physiologie , Protéines virales/métabolisme , Protéines virales/génétique , Coronavirus du syndrome respiratoire du Moyen-Orient/physiologie , Virus du SRAS/physiologie , COVID-19/virologie
10.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-39000573

RÉSUMÉ

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.


Sujet(s)
Protéines de liaison à l'ADN , Mycobactériophages , Mycobacterium smegmatis , Protéines virales , Mycobactériophages/génétique , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Mycobacterium smegmatis/virologie , Mycobacterium smegmatis/génétique , Protéines virales/génétique , Protéines virales/métabolisme , Protéines virales/composition chimique , Lysogénie/génétique , Génome viral , ADN viral/génétique
11.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-39000098

RÉSUMÉ

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.


Sujet(s)
Protéines de capside , Nicotiana , Maladies des plantes , Potexvirus , Solanum tuberosum , Protéines de capside/génétique , Protéines de capside/métabolisme , Nicotiana/génétique , Nicotiana/virologie , Nicotiana/métabolisme , Potexvirus/génétique , Potexvirus/pathogénicité , Maladies des plantes/virologie , Maladies des plantes/génétique , Solanum tuberosum/virologie , Solanum tuberosum/génétique , Solanum tuberosum/métabolisme , Protéines virales/génétique , Protéines virales/métabolisme
12.
Science ; 385(6704): 105-112, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38963841

RÉSUMÉ

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.


Sujet(s)
Endonucleases , Introns , Phages de Pseudomonas , Pseudomonas aeruginosa , Interférence virale , Protéines virales , Endonucleases/métabolisme , Endonucleases/génétique , Interférence virale/génétique , Protéines virales/génétique , Protéines virales/métabolisme , Assemblage viral , Réplication virale , Phages de Pseudomonas/enzymologie , Phages de Pseudomonas/génétique , Pseudomonas aeruginosa/virologie
13.
J Med Virol ; 96(7): e29752, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38949191

RÉSUMÉ

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.


Sujet(s)
COVID-19 , Mitochondries , SARS-CoV-2 , Protéines virales , Humains , Cellules A549 , COVID-19/métabolisme , COVID-19/virologie , COVID-19/anatomopathologie , Mitochondries/métabolisme , Cadres ouverts de lecture , SARS-CoV-2/génétique , Transcriptome , Protéines virales/génétique , Protéines virales/métabolisme , Protéines virales régulatrices ou accessoires/métabolisme , Protéines virales régulatrices ou accessoires/génétique , Protéines viroporines/métabolisme
14.
J Virol ; 98(7): e0049824, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38953667

RÉSUMÉ

Coxsackievirus B3 (CVB3) encodes proteinases that are essential for processing of the translated viral polyprotein. Viral proteinases also target host proteins to manipulate cellular processes and evade innate antiviral responses to promote replication and infection. While some host protein substrates of the CVB3 3C and 2A cysteine proteinases have been identified, the full repertoire of targets is not known. Here, we utilize an unbiased quantitative proteomics-based approach termed terminal amine isotopic labeling of substrates (TAILS) to conduct a global analysis of CVB3 protease-generated N-terminal peptides in both human HeLa and mouse cardiomyocyte (HL-1) cell lines infected with CVB3. We identified >800 proteins that are cleaved in CVB3-infected HeLa and HL-1 cells including the viral polyprotein, known substrates of viral 3C proteinase such as PABP, DDX58, and HNRNPs M, K, and D and novel cellular proteins. Network and GO-term analysis showed an enrichment in biological processes including immune response and activation, RNA processing, and lipid metabolism. We validated a subset of candidate substrates that are cleaved under CVB3 infection and some are direct targets of 3C proteinase in vitro. Moreover, depletion of a subset of TAILS-identified target proteins decreased viral yield. Characterization of two target proteins showed that expression of 3Cpro-targeted cleaved fragments of emerin and aminoacyl-tRNA synthetase complex-interacting multifunctional protein 2 modulated autophagy and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway, respectively. The comprehensive identification of host proteins targeted during virus infection provides insights into the cellular pathways manipulated to facilitate infection. IMPORTANCE: RNA viruses encode proteases that are responsible for processing viral proteins into their mature form. Viral proteases also target and cleave host cellular proteins; however, the full catalog of these target proteins is incomplete. We use a technique called terminal amine isotopic labeling of substrates (TAILS), an N-terminomics to identify host proteins that are cleaved under virus infection. We identify hundreds of cellular proteins that are cleaved under infection, some of which are targeted directly by viral protease. Revealing these target proteins provides insights into the host cellular pathways and antiviral signaling factors that are modulated to promote virus infection and potentially leading to virus-induced pathogenesis.


Sujet(s)
Infections à virus coxsackie , Entérovirus humain B , Protéolyse , Entérovirus humain B/métabolisme , Humains , Souris , Animaux , Cellules HeLa , Infections à virus coxsackie/virologie , Infections à virus coxsackie/métabolisme , Protéines virales/métabolisme , Protéomique/méthodes , Interactions hôte-pathogène , Protéases virales 3C/métabolisme , Lignée cellulaire , Protéases virales/métabolisme , Polyprotéines/métabolisme
15.
Methods Mol Biol ; 2824: 105-120, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039409

RÉSUMÉ

The Rift Valley fever virus is one of the bunyaviruses on the WHO's priority list of pathogens that may cause future pandemics. A better understanding of disease progression and viral pathogenesis is urgently needed to develop treatments. The non-structural proteins NSs and NSm of human pathogenic bunyaviruses represent promising therapeutic targets, as they are often key virulence factors. However, their function is still poorly understood, and their structure is yet unknown, mainly because no successful production of these highly complex proteins has been reported. Here we propose a powerful combination of wheat germ cell-free protein synthesis and NMR to study the structure of these proteins and in particular detail cell-free synthesis and lipid reconstitution methods that can be applied to complex membrane proteins.


Sujet(s)
Système acellulaire , Humains , Spectroscopie par résonance magnétique/méthodes , Protéines virales non structurales/métabolisme , Protéines virales non structurales/composition chimique , Triticum/métabolisme , Résonance magnétique nucléaire biomoléculaire/méthodes , Biosynthèse des protéines , Virus de la fièvre de la vallée du Rift , Protéines virales/métabolisme , Protéines virales/composition chimique
16.
Methods Mol Biol ; 2824: 189-202, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039414

RÉSUMÉ

Affinity enrichment coupled with liquid chromatography-tandem mass spectrometry (AE-LC-MS/MS) enables a comprehensive study of virus-host protein-protein interactions in cells and tissues infected with Rift Valley fever virus (RVFV) or ectopically expressing RVFV proteins. Depending on the research question, different experimental setups with carefully chosen controls are needed. Here, we describe the detailed workflow of sample preparation, processing, and cleanup, while also outlining critical points to consider when designing and performing AE-LC-MS/MS experiments.


Sujet(s)
Interactions hôte-pathogène , Protéomique , Virus de la fièvre de la vallée du Rift , Spectrométrie de masse en tandem , Protéomique/méthodes , Spectrométrie de masse en tandem/méthodes , Chromatographie en phase liquide/méthodes , Humains , Protéines virales/métabolisme , Fièvre de la Vallée du Rift/virologie , Fièvre de la Vallée du Rift/métabolisme , Animaux
17.
Methods Mol Biol ; 2824: 335-345, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039421

RÉSUMÉ

Rift Valley fever (RVF) virus is widespread worldwide and poses a severe threat to human life and property. RVF viral polymerase plays a vital role in the replication and transcription of the virus. Here, we describe how to express and purify this polymerase and perform tests for its in vitro activity assays.


Sujet(s)
Virus de la fièvre de la vallée du Rift , Virus de la fièvre de la vallée du Rift/génétique , Saccharomyces cerevisiae/génétique , Protéines virales/métabolisme , Protéines virales/génétique , Dosages enzymatiques/méthodes , Humains , Expression des gènes , Réplication virale/génétique
18.
Microbiology (Reading) ; 170(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39042422

RÉSUMÉ

DNA manipulation is an essential tool in molecular microbiology research that is dependent on the ability of bacteria to take up and preserve foreign DNA by horizontal gene transfer. This process can be significantly impaired by the activity of bacterial restriction modification systems; bacterial operons comprising paired enzymatic activities that protectively methylate host DNA, while cleaving incoming unmodified foreign DNA. Ocr is a phage-encoded protein that inhibits Type I restriction modification systems, the addition of which significantly improves bacterial transformation efficiency. We recently established an improved and highly efficient transformation protocol for the important human pathogen group A Streptococcus using commercially available recombinant Ocr protein, manufacture of which has since been discontinued. In order to ensure the continued availability of Ocr protein within the research community, we have generated tools and methods for in-house Ocr production and validated the activity of the purified recombinant protein.


Sujet(s)
Protéines recombinantes , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Protéines recombinantes/isolement et purification , Protéines virales/génétique , Protéines virales/métabolisme , Bactériophages/génétique , Bactériophages/enzymologie , Streptococcus pyogenes/génétique , Streptococcus pyogenes/enzymologie , Streptococcus pyogenes/métabolisme , Transformation bactérienne , Type I site-specific deoxyribonuclease/métabolisme , Type I site-specific deoxyribonuclease/génétique , Expression des gènes , Escherichia coli/génétique , Escherichia coli/métabolisme
19.
Front Cell Infect Microbiol ; 14: 1418168, 2024.
Article de Anglais | MEDLINE | ID: mdl-38988816

RÉSUMÉ

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.


Sujet(s)
Complexes de tri endosomique requis pour le transport , Exosomes , Interactions hôte-pathogène , microARN , Maladies virales , Exosomes/métabolisme , Humains , Complexes de tri endosomique requis pour le transport/métabolisme , Maladies virales/métabolisme , Maladies virales/virologie , microARN/métabolisme , microARN/génétique , Animaux , Virus/pathogénicité , Virus/métabolisme , Libération de particules virales , Protéines virales/métabolisme , Protéines virales/génétique
20.
PLoS Pathog ; 20(7): e1012345, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38968329

RÉSUMÉ

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.


Sujet(s)
Systèmes CRISPR-Cas , Grippe humaine , Stabilité de l'ARN , ARN messager , ARN viral , Animaux , ARN viral/génétique , ARN viral/métabolisme , ARN messager/génétique , ARN messager/métabolisme , Humains , Grippe humaine/virologie , Sous-type H1N1 du virus de la grippe A/génétique , Sous-type H3N2 du virus de la grippe A/génétique , Infections à Orthomyxoviridae/virologie , Antiviraux/pharmacologie , Chiens , Cricetinae , Protéines virales/génétique , Protéines virales/métabolisme , Mesocricetus , Cellules rénales canines Madin-Darby
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE