Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 78.210
Filtrer
1.
J Med Virol ; 96(7): e29783, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38965890

RÉSUMÉ

Many COVID-19 patients suffer from gastrointestinal symptoms and impaired intestinal barrier function is thought to play a key role in Long COVID. Despite its importance, the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on intestinal epithelia is poorly understood. To address this, we established an intestinal barrier model integrating epithelial Caco-2 cells, mucus-secreting HT29 cells and Raji cells. This gut epithelial model allows efficient differentiation of Caco-2 cells into microfold-like cells, faithfully mimics intestinal barrier function, and is highly permissive to SARS-CoV-2 infection. Early strains of SARS-CoV-2 and the Delta variant replicated with high efficiency, severely disrupted barrier function, and depleted tight junction proteins, such as claudin-1, occludin, and ZO-1. In comparison, Omicron subvariants also depleted ZO-1 from tight junctions but had fewer damaging effects on mucosal integrity and barrier function. Remdesivir, the fusion inhibitor EK1 and the transmembrane serine protease 2 inhibitor Camostat inhibited SARS-CoV-2 replication and thus epithelial barrier damage, while the Cathepsin inhibitor E64d was ineffective. Our results support that SARS-CoV-2 disrupts intestinal barrier function but further suggest that circulating Omicron variants are less damaging than earlier viral strains.


Sujet(s)
COVID-19 , Muqueuse intestinale , SARS-CoV-2 , Jonctions serrées , Réplication virale , Humains , SARS-CoV-2/pathogénicité , Cellules Caco-2 , COVID-19/virologie , COVID-19/anatomopathologie , Muqueuse intestinale/virologie , Muqueuse intestinale/anatomopathologie , Jonctions serrées/virologie , Alanine/analogues et dérivés , Protéine-1 de la zonula occludens/métabolisme , Protéine-1 de la zonula occludens/génétique , Antiviraux/pharmacologie , Cellules HT29 , Occludine/métabolisme , Occludine/génétique , AMP/analogues et dérivés
2.
BMC Vet Res ; 20(1): 288, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961481

RÉSUMÉ

BACKGROUND: Porcine epidemic diarrhea virus (PEDV) mainly causes acute and severe porcine epidemic diarrhea (PED), and is highly fatal in neonatal piglets. No reliable therapeutics against the infection exist, which poses a major global health issue for piglets. Luteolin is a flavonoid with anti-viral activity toward several viruses. RESULTS: We evaluated anti-viral effects of luteolin in PEDV-infected Vero and IPEC-J2 cells, and identified IC50 values of 23.87 µM and 68.5 µM, respectively. And found PEDV internalization, replication and release were significantly reduced upon luteolin treatment. As luteolin could bind to human ACE2 and SARS-CoV-2 main protease (Mpro) to contribute viral entry, we first identified that luteolin shares the same core binding site on pACE2 with PEDV-S by molecular docking and exhibited positive pACE2 binding with an affinity constant of 71.6 µM at dose-dependent increases by surface plasmon resonance (SPR) assay. However, pACE2 was incapable of binding to PEDV-S1. Therefore, luteolin inhibited PEDV internalization independent of PEDV-S binding to pACE2. Moreover, luteolin was firmly embedded in the groove of active pocket of Mpro in a three-dimensional docking model, and fluorescence resonance energy transfer (FRET) assays confirmed that luteolin inhibited PEDV Mpro activity. In addition, we also observed PEDV-induced pro-inflammatory cytokine inhibition and Nrf2-induced HO-1 expression. Finally, a drug resistant mutant was isolated after 10 cell culture passages concomitant with increasing luteolin concentrations, with reduced PEDV susceptibility to luteolin identified at passage 10. CONCLUSIONS: Our results push forward that anti-PEDV mechanisms and resistant-PEDV properties for luteolin, which may be used to combat PED.


Sujet(s)
Antiviraux , Lutéoline , Virus de la diarrhée porcine épidémique , Lutéoline/pharmacologie , Virus de la diarrhée porcine épidémique/effets des médicaments et des substances chimiques , Animaux , Antiviraux/pharmacologie , Chlorocebus aethiops , Cellules Vero , Suidae , Simulation de docking moléculaire , Pénétration virale/effets des médicaments et des substances chimiques , Réplication virale/effets des médicaments et des substances chimiques , Lignée cellulaire , Simulation numérique , Maladies des porcs/virologie , Maladies des porcs/traitement médicamenteux
3.
Front Immunol ; 15: 1424374, 2024.
Article de Anglais | MEDLINE | ID: mdl-38966641

RÉSUMÉ

At the beginning of the COVID-19 pandemic those with underlying chronic lung conditions, including tuberculosis (TB), were hypothesized to be at higher risk of severe COVID-19 disease. However, there is inconclusive clinical and preclinical data to confirm the specific risk SARS-CoV-2 poses for the millions of individuals infected with Mycobacterium tuberculosis (M.tb). We and others have found that compared to singly infected mice, mice co-infected with M.tb and SARS-CoV-2 leads to reduced SARS-CoV-2 severity compared to mice infected with SARS-CoV-2 alone. Consequently, there is a large interest in identifying the molecular mechanisms responsible for the reduced SARS-CoV-2 infection severity observed in M.tb and SARS-CoV-2 co-infection. To address this, we conducted a comprehensive characterization of a co-infection model and performed mechanistic in vitro modeling to dynamically assess how the innate immune response induced by M.tb restricts viral replication. Our study has successfully identified several cytokines that induce the upregulation of anti-viral genes in lung epithelial cells, thereby providing protection prior to challenge with SARS-CoV-2. In conclusion, our study offers a comprehensive understanding of the key pathways induced by an existing bacterial infection that effectively restricts SARS-CoV-2 activity and identifies candidate therapeutic targets for SARS-CoV-2 infection.


Sujet(s)
COVID-19 , Co-infection , Immunité innée , Mycobacterium tuberculosis , SARS-CoV-2 , COVID-19/immunologie , Animaux , Mycobacterium tuberculosis/immunologie , SARS-CoV-2/immunologie , SARS-CoV-2/physiologie , Souris , Co-infection/immunologie , Humains , Tuberculose/immunologie , Tuberculose/microbiologie , Cytokines/métabolisme , Cytokines/immunologie , Modèles animaux de maladie humaine , Indice de gravité de la maladie , Poumon/immunologie , Poumon/virologie , Poumon/microbiologie , Poumon/anatomopathologie , Réplication virale , Souris de lignée C57BL , Femelle
4.
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
5.
J Nanobiotechnology ; 22(1): 388, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38956618

RÉSUMÉ

BACKGROUND: Porcine reproductive and respiratory syndrome virus (PRRSV) is a prevalent swine pathogen, which has caused adverse impact on the global swine industry for almost 30 years. However, due to the immune suppression caused by the virus and the genetic diversity in PRRSV, no virus-targeting broad neutralizing strategy has been successfully developed yet. Antiviral peptide and nanobody have attracted extensive attention with the ease in production and the efficacy in practice. In this study, four new fusion proteins named nanobody peptide conjugates (NPCs) were developed by combining PRRSV specific non-neutralizing nanobodies with CD163-derived peptides targeting the receptor binding domain (RBD) of PRRSV proteins. RESULTS: Four NPCs were successfully constructed using two nanobodies against PRRSV N and nsp9 individually, recombining with two antiviral peptides 4H7 or 8H2 from porcine CD163 respectively. All four NPCs demonstrated specific capability of binding to PRRSV and broad inhibitory effect against various lineages of PRRSV in a dose-dependent manner. NPCs interfere with the binding of the RBD of PRRSV proteins to CD163 in the PRRSV pre-attachment stage by CD163 epitope peptides in the assistance of Nb components. NPCs also suppress viral replication during the stage of post-attachment, and the inhibitory effects depend on the antiviral functions of Nb parts in NPCs, including the interference in long viral RNA synthesis, NF-κB and IFN-ß activation. Moreover, an interaction was predicted between aa K31 and T32 sites of neutralizing domain 4H7 of NPC-N/nsp9-4H7 and the motif 171NLRLTG176 of PRRSV GP2a. The motif 28SSS30 of neutralizing domain 8H2 of NPC-N/nsp9-8H2 could also form hydrogens to bind with the motif 152NAFLP156 of PRRSV GP3. The study provides valuable insights into the structural characteristics and potential functional implications of the RBD of PRRSV proteins. Finally, as indicated in a mouse model, NPC intranasally inoculated in vivo for 12-24 h sustains the significant neutralizing activity against PRRSV. These findings inspire the potential of NPC as a preventive measure to reduce the transmission risk in the host population against respiratory infectious agents like PRRSV. CONCLUSION: The aim of the current study was to develop a peptide based bioactive compound to neutralize various PRRSV strains. The new antiviral NPC (nanobody peptide conjugate) consists of a specific nanobody targeting the viral protein and a neutralizing CD163 epitope peptide for virus blocking and provides significant antiviral activity. The study will greatly promote the antiviral drug R&D against PRRSV and enlighten a new strategy against other viral diseases.


Sujet(s)
Anticorps neutralisants , Antigènes CD , Antigènes de différenciation des myélomonocytes , Peptides , Virus du syndrome respiratoire et reproducteur porcin , Récepteurs de surface cellulaire , Anticorps à domaine unique , Virus du syndrome respiratoire et reproducteur porcin/immunologie , Virus du syndrome respiratoire et reproducteur porcin/effets des médicaments et des substances chimiques , Animaux , Anticorps à domaine unique/immunologie , Anticorps à domaine unique/pharmacologie , Anticorps à domaine unique/composition chimique , Suidae , Antigènes de différenciation des myélomonocytes/immunologie , Antigènes de différenciation des myélomonocytes/métabolisme , Récepteurs de surface cellulaire/immunologie , Antigènes CD/immunologie , Antigènes CD/métabolisme , Anticorps neutralisants/immunologie , Peptides/composition chimique , Peptides/pharmacologie , Peptides/immunologie , Syndrome dysgénésique et respiratoire porcin/immunologie , Syndrome dysgénésique et respiratoire porcin/prévention et contrôle , Souris , Réplication virale/effets des médicaments et des substances chimiques , Lignée cellulaire
6.
PLoS Biol ; 22(7): e3002696, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38959200

RÉSUMÉ

Sterile alpha motif domain-containing proteins 9 and 9-like (SAMD9/9L) are associated with life-threatening genetic diseases in humans and are restriction factors of poxviruses. Yet, their cellular function and the extent of their antiviral role are poorly known. Here, we found that interferon-stimulated human SAMD9L restricts HIV-1 in the late phases of replication, at the posttranscriptional and prematuration steps, impacting viral translation and, possibly, endosomal trafficking. Surprisingly, the paralog SAMD9 exerted an opposite effect, enhancing HIV-1. More broadly, we showed that SAMD9L restricts primate lentiviruses, but not a gammaretrovirus (MLV), nor 2 RNA viruses (arenavirus MOPV and rhabdovirus VSV). Using structural modeling and mutagenesis of SAMD9L, we identified a conserved Schlafen-like active site necessary for HIV-1 restriction by human and a rodent SAMD9L. By testing a gain-of-function constitutively active variant from patients with SAMD9L-associated autoinflammatory disease, we determined that SAMD9L pathogenic functions also depend on the Schlafen-like active site. Finally, we found that the constitutively active SAMD9L strongly inhibited HIV, MLV, and, to a lesser extent, MOPV. This suggests that the virus-specific effect of SAMD9L may involve its differential activation/sensing and the virus ability to evade from SAMD9L restriction. Overall, our study identifies SAMD9L as an HIV-1 antiviral factor from the cell autonomous immunity and deciphers host determinants underlying the translational repression. This provides novel links and therapeutic avenues against viral infections and genetic diseases.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Lentivirus des primates , Réplication virale , Humains , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Animaux , Lentivirus des primates/génétique , Lentivirus des primates/métabolisme , Cellules HEK293 , Biosynthèse des protéines , Facteurs de restriction antiviraux , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Infections à VIH/virologie , Infections à VIH/traitement médicamenteux , Protéines suppresseurs de tumeurs
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 Vis Exp ; (208)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38949380

RÉSUMÉ

Viral infections can cause Endoplasmic Reticulum (ER) stress due to abnormal protein accumulation, leading to Unfolded Protein Response (UPR). Viruses have developed strategies to manipulate the host UPR, but there is a lack of detailed understanding of UPR modulation and its functional significance during HIV-1 infection in the literature. In this context, the current article describes the protocols used in our laboratory to measure ER stress levels and UPR during HIV-1 infection in T-cells and the effect of UPR on viral replication and infectivity. Thioflavin T (ThT) staining is a relatively new method used to detect ER stress in the cells by detecting protein aggregates. Here, we have illustrated the protocol for ThT staining in HIV-1 infected cells to detect and quantify ER stress. Moreover, ER stress was also detected indirectly by measuring the levels of UPR markers such as BiP, phosphorylated IRE1, PERK, and eIF2α, splicing of XBP1, cleavage of ATF6, ATF4, CHOP, and GADD34 in HIV-1 infected cells, using conventional immunoblotting and quantitative reverse transcription polymerase chain reaction (RT-PCR). We have found that the ThT-fluorescence correlates with the indicators of UPR activation. This article also demonstrates the protocols to analyze the impact of ER stress and UPR modulation on HIV-1 replication by knockdown experiments as well as the use of pharmacological molecules. The effect of UPR on HIV-1 gene expression/replication and virus production was analyzed by Luciferase reporter assays and p24 antigen capture ELISA, respectively, whereas the effect on virion infectivity was analyzed by staining of infected reporter cells. Collectively, this set of methods provides a comprehensive understanding of the Unfolded Protein Response pathways during HIV-1 infection, revealing its intricate dynamics.


Sujet(s)
Stress du réticulum endoplasmique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Réponse aux protéines mal repliées , Réplication virale , Humains , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Réplication virale/physiologie , Stress du réticulum endoplasmique/physiologie , Infections à VIH/virologie , Infections à VIH/métabolisme , Lymphocytes T/virologie , Lymphocytes T/métabolisme
10.
Environ Microbiol ; 26(7): e16670, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38952172

RÉSUMÉ

The influence of environmental factors on the interactions between phages and bacteria, particularly single-stranded DNA (ssDNA) phages, has been largely unexplored. In this study, we used Finnlakevirus FLiP, the first known ssDNA phage species with a lipid membrane, as our model phage. We examined the infectivity of FLiP with three Flavobacterium host strains, B330, B167 and B114. We discovered that FLiP infection is contingent on the host strain and conditions such as temperature and bacterial growth phase. FLiP can infect its hosts across a wide temperature range, but optimal phage replication varies with each host. We uncovered some unique aspects of phage infectivity: FLiP has limited infectivity in liquid-suspended cells, but it improves when cells are surface-attached. Moreover, FLiP infects stationary phase B167 and B114 cells more rapidly and efficiently than exponentially growing cells, a pattern not observed with the B330 host. We also present the first experimental evidence of endolysin function in ssDNA phages. The activity of FLiP's lytic enzymes was found to be condition-dependent. Our findings underscore the importance of studying phage ecology in contexts that are relevant to the environment, as both the host and the surrounding conditions can significantly alter the outcome of phage-host interactions.


Sujet(s)
Bactériophages , ADN simple brin , Flavobacterium , ADN simple brin/métabolisme , ADN simple brin/génétique , Bactériophages/génétique , Bactériophages/physiologie , Flavobacterium/virologie , Flavobacterium/croissance et développement , Flavobacterium/génétique , Interactions hôte-microbes , Endopeptidases/métabolisme , Endopeptidases/génétique , Réplication virale , Température
11.
Arch Insect Biochem Physiol ; 116(3): e22127, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38976652

RÉSUMÉ

Ubiquitin-fold modifier 1 (UFM1) is attached to protein substrates through the sequential activity of an E1 (UBA5)-E2 (UFC1)-E3 (UFL1) cascade. UFL1 is the E3 ligase for UFMylation in vertebrates. However, there have been no studies on UFL1 in silkworm to date. In this study, we identified a UFL1 ortholog in Bombyx mori genome. Spatio-temporal expression profiles showed that BmUFL1 expression was high in the midgut, epidermis, and testis and in the pupa-adult stage. BmUFL1 knockdown inhibited B. mori nucleopolyhedrovirus (BmNPV) proliferation, while BmUFL1 overexpression promoted BmNPV proliferation. Mechanically, protein kinase RNA-like endoplasmic reticulum kinase (PERK) signaling and cell apoptosis are involved in BmUFL1-regulated BmNPV proliferation. Overall, these results suggest that BmUFL1 facilitates BmNPV proliferation in silkworm.


Sujet(s)
Apoptose , Bombyx , Protéines d'insecte , Nucleopolyhedrovirus , eIF-2 Kinase , Animaux , Bombyx/virologie , Bombyx/génétique , Bombyx/croissance et développement , Nucleopolyhedrovirus/physiologie , Protéines d'insecte/métabolisme , Protéines d'insecte/génétique , eIF-2 Kinase/métabolisme , eIF-2 Kinase/génétique , Réplication virale , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Larve/virologie , Larve/croissance et développement , Larve/métabolisme , Larve/génétique
12.
Arch Microbiol ; 206(8): 345, 2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-38976047

RÉSUMÉ

Neurological complications, both acute and chronic, are reported commonly in COVID-19 affected individuals. In this context, the understanding of pathogenesis of SARS-CoV-2 in specific cells of central nervous system (CNS) origin is relevant. The present study explores infection biology of a clinical isolate of SARS-CoV-2 in human cell lines of neural origin such as the glioblastoma (U87-MG), neuroblastoma (SHSY5Y) and microglia (C20). Despite showing clear evidence of infection by immunofluorescence with an anti-spike protein antibody, all the three neural cell lines were observed to be highly restrictive to the replication of the infecting virus. While the U87-MG glioblastoma cells demonstrated no cytopathic effects and a low viral titre with no signs of replication, the SHSY5Y neuroblastoma cells exhibited cytopathic effects with bleb formation but no evidence of viable virus. The C20 microglial cells showed neither signs of cytopathic effects nor viable virus. Ultrastructural studies demonstrated intracellular virions in infected neural cells. The presence of lipid droplets in infected SHSY5Y cells suggested an impact on host cell metabolism. The decrease in viral RNA levels over time in all the neural cell lines suggested restricted viral replication. In conclusion, this study highlights the limited susceptibility of neural cells to SARS-CoV-2 infection. This reduced permissibility of neural cell lines to SARS-CoV-2 may point to their inherent lower expression of receptors that support viral entry in addition to the intracellular factors that potently inhibit viral replication. The study findings prompt further investigation into the mechanisms of SARS-CoV-2 infection of neural cells.


Sujet(s)
COVID-19 , Microglie , Névroglie , Neurones , SARS-CoV-2 , Réplication virale , Humains , Microglie/virologie , SARS-CoV-2/physiologie , SARS-CoV-2/pathogénicité , Neurones/virologie , COVID-19/virologie , Névroglie/virologie , Lignée cellulaire tumorale , Lignée cellulaire , Effet cytopathogène viral , Glycoprotéine de spicule des coronavirus/métabolisme , ARN viral/génétique
13.
Virologie (Montrouge) ; 28(3): 199-215, 2024 Jun 01.
Article de Français | MEDLINE | ID: mdl-38970341

RÉSUMÉ

Viroids are the smallest non-coding infectious RNAs (between 246 and 401 nucleotides) known to be highly structured and replicate autonomously in the host plants. Although they do not encode any peptides, viroids induce visible symptoms in susceptible host plants. This article provides an overview of their physical and biological properties, the diseases they cause and their significance for the plants. The mechanisms underlying the expression of symptoms in host plants, their detection and various strategies employed for diseases prevention are also developed.


Sujet(s)
Maladies des plantes , Plantes , ARN viral , Viroïdes , Viroïdes/génétique , Viroïdes/physiologie , Maladies des plantes/virologie , Maladies des plantes/prévention et contrôle , ARN viral/génétique , ARN non traduit/génétique , ARN non traduit/physiologie , Réplication virale
14.
Virologie (Montrouge) ; 28(3): 187-197, 2024 Jun 01.
Article de Français | MEDLINE | ID: mdl-38970340

RÉSUMÉ

Orthoflaviviruses are enveloped positive-sense RNA viruses comprising numerous human pathogens transmitted by hematophagous arthropods. This includes viruses such as dengue virus, Zika virus, and yellow fever virus. The viral nonstructural protein NS1 plays a central role in the pathogenesis and cycle of these viruses by acting in two different forms: associated with the plasma membrane (NS1m) or secreted outside the cell (NS1s). The versatility of NS1 is evident in its ability to modulate various aspects of the infectious process, from immune evasion to pathogenesis. As an intracellular protein, it disrupts many processes, interfering with signaling pathways and facilitating viral replication in concert with other viral proteins. As a secreted protein, NS1 actively participates in immune evasion, interfering with the host immune system, inhibiting the complement system, facilitating viral dissemination, and disrupting the integrity of endothelial barriers. This review primarily aims to address the role of NS1 in viral pathogenesis associated with orthoflaviviruses.


Sujet(s)
Protéines virales non structurales , Réplication virale , Protéines virales non structurales/métabolisme , Protéines virales non structurales/physiologie , Humains , Animaux , Infections à flavivirus/virologie , Échappement immunitaire , Flavivirus/physiologie , Flavivirus/pathogénicité , Virus Zika/physiologie , Virus Zika/pathogénicité , Virus de la dengue/physiologie
15.
Vet Microbiol ; 295: 110163, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38959807

RÉSUMÉ

Avian influenza virus (AIV) infection and vaccination against live attenuated infectious bronchitis virus (aIBV) are frequent in poultry worldwide. Here, we evaluated the clinical effect of H9N2 subtype AIV and QX genotype aIBV co-infection in specific-pathogen-free (SPF) white leghorn chickens and explored the potential mechanisms underlying the observed effects using by 4D-FastDIA-based proteomics. The results showed that co-infection of H9N2 AIV and QX aIBV increased mortality and suppressed the growth of SPF chickens. In particular, severe lesions in the kidneys and slight respiratory signs similar to the symptoms of virulent QX IBV infection were observed in some co-infected chickens, with no such clinical signs observed in single-infected chickens. The replication of H9N2 AIV was significantly enhanced in both the trachea and kidneys, whereas there was only a slight effect on the replication of the QX aIBV. Proteomics analysis showed that the IL-17 signaling pathway was one of the unique pathways enriched in co-infected chickens compared to single infected-chickens. A series of metabolism and immune response-related pathways linked with co-infection were also significantly enriched. Moreover, co-infection of the two pathogens resulted in the enrichment of the negative regulation of telomerase activity. Collectively, our study supports the synergistic effect of the two pathogens, and pointed out that aIBV vaccines might increased IBV-associated lesions due to pathogenic co-infections. Exacerbation of the pathogenicity and mortality in H9N2 AIV and QX aIBV co-infected chickens possibly occurred because of an increase in H9N2 AIV replication, the regulation of telomerase activity, and the disturbance of cell metabolism and the immune system.


Sujet(s)
Poulets , Co-infection , Infections à coronavirus , Virus de la bronchite infectieuse , Sous-type H9N2 du virus de la grippe A , Grippe chez les oiseaux , Maladies de la volaille , Animaux , Poulets/virologie , Sous-type H9N2 du virus de la grippe A/pathogénicité , Sous-type H9N2 du virus de la grippe A/génétique , Virus de la bronchite infectieuse/pathogénicité , Virus de la bronchite infectieuse/génétique , Co-infection/virologie , Co-infection/médecine vétérinaire , Grippe chez les oiseaux/virologie , Maladies de la volaille/virologie , Infections à coronavirus/médecine vétérinaire , Infections à coronavirus/virologie , Organismes exempts d'organismes pathogènes spécifiques , Réplication virale , Vaccins atténués/immunologie , Génotype , Virulence , Protéomique , Rein/virologie , Rein/anatomopathologie
16.
Sci Rep ; 14(1): 15351, 2024 07 04.
Article de Anglais | MEDLINE | ID: mdl-38961189

RÉSUMÉ

As SARS-CoV-2 continues to spread worldwide, tractable primary airway cell models that recapitulate the cell-intrinsic response to arising viral variants are needed. Here we describe an adult stem cell-derived human airway organoid model overexpressing the ACE2 receptor (ACE2-OE) that supports robust viral replication while maintaining 3D architecture and cellular diversity of the airway epithelium. ACE2-OE organoids were infected with SARS-CoV-2 variants and subjected to single-cell RNA-sequencing. Interferon-lambda was upregulated in cells with low-level infection while the NF-kB inhibitor alpha gene (encoding IkBa) was consistently upregulated in infected cells, and its expression positively correlated with infection levels. Confocal microscopy showed more IkBa expression in infected than bystander cells, but found concurrent nuclear translocation of NF-kB that IkBa usually prevents. Overexpressing a nondegradable IkBa mutant reduced NF-kB translocation and increased viral infection. These data demonstrate the functionality of ACE2-OE organoids in SARS-CoV-2 research and underscore that the strength of the NF-kB feedback loop in infected cells controls viral replication.


Sujet(s)
Angiotensin-converting enzyme 2 , COVID-19 , Inhibiteur alpha de NF-KappaB , Organoïdes , SARS-CoV-2 , Réplication virale , Humains , Organoïdes/virologie , Organoïdes/métabolisme , Angiotensin-converting enzyme 2/métabolisme , Angiotensin-converting enzyme 2/génétique , SARS-CoV-2/physiologie , COVID-19/virologie , COVID-19/métabolisme , COVID-19/génétique , Inhibiteur alpha de NF-KappaB/métabolisme , Inhibiteur alpha de NF-KappaB/génétique , Facteur de transcription NF-kappa B/métabolisme
17.
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
18.
Arch Virol ; 169(8): 164, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38990242

RÉSUMÉ

Upregulation of ADAMTS-4 has been reported to have an important role in lung injury, and ADAMTS-4 expression is regulated by miR-126a-5p in abdominal aortic aneurysms. The aim of this study was to investigate whether miR-126a-5p/ADAMTS-4 plays a role in influenza-virus-induced lung injury. Lung fibroblasts were infected with H1N1 influenza virus to detect changes in miR-126a-5p and ADAMTS-4 expression, and cell viability was measured by CCK-8 assay. Inflammatory factors and matrix protease levels were examined using ELISA kits, and cell apoptosis was assessed by measuring the levels of apoptosis-related proteins. A dual luciferase assay was used to verify the regulatory relationship between miR-126a-5p and ADAMTS-4. H1N1 influenza virus reduced fibroblast viability, inhibited miR-126a-5p expression, and promoted ADAMTS-4 expression. Overexpression of miR-126a-5p attenuated the cellular inflammatory response, apoptosis, matrix protease secretion, and virus replication. Luciferase reporter assays revealed that miR-126a-5p inhibited ADAMTS-4 expression by targeting ADAMTS-4 mRNA. Further experiments showed that overexpression of ADAMTS-4 significantly reversed the inhibitory effects of miR-126a-5p on fibroblast inflammation, apoptosis, matrix protease secretion, and virus replication. Upregulation of miR-126a-5p inhibits H1N1-induced apoptosis, inflammatory factors, and matrix protease secretion, as well as virus replication in lung fibroblasts.


Sujet(s)
Protéine ADAMTS4 , Apoptose , Fibroblastes , Inflammation , Sous-type H1N1 du virus de la grippe A , Poumon , microARN , microARN/génétique , microARN/métabolisme , Fibroblastes/virologie , Fibroblastes/métabolisme , Humains , Poumon/virologie , Poumon/anatomopathologie , Protéine ADAMTS4/génétique , Protéine ADAMTS4/métabolisme , Sous-type H1N1 du virus de la grippe A/génétique , Sous-type H1N1 du virus de la grippe A/physiologie , Inflammation/génétique , Survie cellulaire , Réplication virale , Grippe humaine/virologie , Grippe humaine/génétique , Grippe humaine/métabolisme , Lignée cellulaire
19.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-39000089

RÉSUMÉ

In the field of virology, liquid-liquid phase separation (LLPS) has emerged as a pivotal mechanism enabling the compartmentalization required for specific steps of the viral replication cycle [...].


Sujet(s)
Maladies virales , Humains , Maladies virales/virologie , Réplication virale , Extraction liquide-liquide/méthodes ,
20.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000271

RÉSUMÉ

The human immunodeficiency virus type 1 (HIV-1) capsid is a protein core formed by multiple copies of the viral capsid (CA) protein. Inside the capsid, HIV-1 harbours all the viral components required for replication, including the genomic RNA and viral enzymes reverse transcriptase (RT) and integrase (IN). Upon infection, the RT transforms the genomic RNA into a double-stranded DNA molecule that is subsequently integrated into the host chromosome by IN. For this to happen, the viral capsid must open and release the viral DNA, in a process known as uncoating. Capsid plays a key role during the initial stages of HIV-1 replication; therefore, its stability is intimately related to infection efficiency, and untimely uncoating results in reverse transcription defects. How and where uncoating takes place and its relationship with reverse transcription is not fully understood, but the recent development of novel biochemical and cellular approaches has provided unprecedented detail on these processes. In this review, we present the latest findings on the intricate link between capsid stability, reverse transcription and uncoating, the different models proposed over the years for capsid uncoating, and the role played by other cellular factors on these processes.


Sujet(s)
Protéines de capside , Capside , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Transcription inverse , Décapsidation virale , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Humains , Capside/métabolisme , Protéines de capside/métabolisme , Protéines de capside/génétique , Réplication virale , Infections à VIH/virologie , Infections à VIH/métabolisme , ARN viral/métabolisme , ARN viral/génétique , Transcriptase inverse du VIH/métabolisme , Transcriptase inverse du VIH/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...