Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.504
Filtrer
1.
Nat Commun ; 15(1): 5923, 2024 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-39004634

RÉSUMÉ

Respiratory syncytial virus (RSV) is an enveloped, filamentous, negative-strand RNA virus that causes significant respiratory illness worldwide. RSV vaccines are available, however there is still significant need for research to support the development of vaccines and therapeutics against RSV and related Mononegavirales viruses. Individual virions vary in size, with an average diameter of ~130 nm and ranging from ~500 nm to over 10 µm in length. Though the general arrangement of structural proteins in virions is known, we use cryo-electron tomography and sub-tomogram averaging to determine the molecular organization of RSV structural proteins. We show that the peripheral membrane-associated RSV matrix (M) protein is arranged in a packed helical-like lattice of M-dimers. We report that RSV F glycoprotein is frequently observed as pairs of trimers oriented in an anti-parallel conformation to support potential interactions between trimers. Our sub-tomogram averages indicate the positioning of F-trimer pairs is correlated with the underlying M lattice. These results provide insight into RSV virion organization and may aid in the development of RSV vaccines and anti-viral targets.


Sujet(s)
Cryomicroscopie électronique , Virus respiratoire syncytial humain , Protéines de fusion virale , Protéines de la matrice virale , Protéines de fusion virale/composition chimique , Protéines de fusion virale/métabolisme , Protéines de la matrice virale/composition chimique , Protéines de la matrice virale/métabolisme , Protéines de la matrice virale/ultrastructure , Humains , Virus respiratoire syncytial humain/composition chimique , Multimérisation de protéines , Virion/métabolisme , Virion/ultrastructure , Virion/composition chimique , Tomographie en microscopie électronique , Virus respiratoires syncytiaux/composition chimique , Modèles moléculaires , Infections à virus respiratoire syncytial/virologie , Animaux
2.
Nat Commun ; 15(1): 5514, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951492

RÉSUMÉ

HIV-1 Vpr promotes efficient spread of HIV-1 from macrophages to T cells by transcriptionally downmodulating restriction factors that target HIV-1 Envelope protein (Env). Here we find that Vpr induces broad transcriptomic changes by targeting PU.1, a transcription factor necessary for expression of host innate immune response genes, including those that target Env. Consistent with this, we find silencing PU.1 in infected macrophages lacking Vpr rescues Env. Vpr downmodulates PU.1 through a proteasomal degradation pathway that depends on physical interactions with PU.1 and DCAF1, a component of the Cul4A E3 ubiquitin ligase. The capacity for Vpr to target PU.1 is highly conserved across primate lentiviruses. In addition to impacting infected cells, we find that Vpr suppresses expression of innate immune response genes in uninfected bystander cells, and that virion-associated Vpr can degrade PU.1. Together, we demonstrate Vpr counteracts PU.1 in macrophages to blunt antiviral immune responses and promote viral spread.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Immunité innée , Macrophages , Protéines proto-oncogènes , Transactivateurs , Produits du gène vpr du virus de l'immunodéficience humaine , Humains , Macrophages/immunologie , Macrophages/métabolisme , Macrophages/virologie , Produits du gène vpr du virus de l'immunodéficience humaine/métabolisme , Produits du gène vpr du virus de l'immunodéficience humaine/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Transactivateurs/métabolisme , Transactivateurs/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes/génétique , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Infections à VIH/immunologie , Infections à VIH/virologie , Infections à VIH/génétique , Cellules HEK293 , Virion/métabolisme , Protein-Serine-Threonine Kinases
3.
J Extracell Vesicles ; 13(7): e12476, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38978287

RÉSUMÉ

The current study analyzed the intersecting biophysical, biochemical, and functional properties of extracellular particles (EPs) with the human immunodeficiency virus type-1 (HIV-1) beyond the currently accepted size range for HIV-1. We isolated five fractions (Frac-A through Frac-E) from HIV-infected cells by sequential differential ultracentrifugation (DUC). All fractions showed a heterogeneous size distribution with median particle sizes greater than 100 nm for Frac-A through Frac-D but not for Frac-E, which contained small EPs with an average size well below 50 nm. Synchronized and released cultures contained large infectious EPs in Frac-A, with markers of amphisomes and viral components. Additionally, Frac-E uniquely contained EPs positive for CD63, HSP70, and HIV-1 proteins. Despite its small average size, Frac-E contained membrane-protected viral integrase, detectable only after SDS treatment, indicating that it is enclosed in vesicles. Single particle analysis with dSTORM further supported these findings as CD63, HIV-1 integrase, and the viral surface envelope (Env) glycoprotein (gp) colocalized on the same Frac-E particles. Surprisingly, Frac-E EPs were infectious, and infectivity was significantly reduced by immunodepleting Frac-E with anti-CD63, indicating the presence of this protein on the surface of infectious small EPs in Frac-E. To our knowledge, this is the first time that extracellular vesicle (EV) isolation methods have identified infectious small HIV-1 particles (smHIV-1) that are under 50 nm. Collectively, our data indicate that the crossroads between EPs and HIV-1 potentially extend beyond the currently accepted biophysical properties of HIV-1, which may have further implications for viral pathogenesis.


Sujet(s)
Vésicules extracellulaires , Infections à VIH , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Humains , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/virologie , Infections à VIH/virologie , Infections à VIH/métabolisme , Virion/métabolisme , Ultracentrifugation/méthodes , Lymphocytes T/virologie , Lymphocytes T/métabolisme , Antigène CD63/métabolisme , Taille de particule
4.
J Virol ; 98(7): e0085024, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38953378

RÉSUMÉ

Viruses are obligate parasites that depend on the cellular machinery for their propagation. Several viruses also incorporate cellular proteins that facilitate viral spread. Defining these cellular proteins is critical to decipher viral life cycles and delineate novel therapeutic strategies. While numerous studies have explored the importance of host proteins in coronavirus spread, information about their presence in mature virions is limited. In this study, we developed a protocol to highly enrich mature HCoV-OC43 virions and characterize them by proteomics. Recognizing that cells release extracellular vesicles whose content is modulated by viruses, and given our ability to separate virions from these vesicles, we also analyzed their protein content in both uninfected and infected cells. We uncovered 69 unique cellular proteins associated with virions including 31 high-confidence hits. These proteins primarily regulate RNA metabolism, enzymatic activities, vesicular transport, cell adhesion, metabolite interconversion, and translation. We further discovered that the virus had a profound impact on exosome composition, incorporating 47 novel cellular proteins (11 high confidence) and excluding 92 others (61 high confidence) in virus-associated extracellular vesicles compared to uninfected cells. Moreover, a dsiRNA screen revealed that 11 of 18 select targets significantly impacted viral yields, including proteins found in virions or extracellular vesicles. Overall, this study provides new and important insights into the incorporation of numerous host proteins into HCoV-OC43 virions, their biological significance, and the ability of the virus to modulate extracellular vesicles. IMPORTANCE: In recent years, coronaviruses have dominated global attention, making it crucial to develop methods to control them and prevent future pandemics. Besides viral proteins, host proteins play a significant role in viral propagation and offer potential therapeutic targets. Targeting host proteins is advantageous because they are less likely to mutate and develop resistance compared to viral proteins, a common issue with many antiviral treatments. In this study, we examined the protein content of the less virulent biosafety level 2 HCoV-OC43 virus as a stand-in for the more virulent SARS-CoV-2. Our findings reveal that several cellular proteins incorporated into the virion regulate viral spread. In addition, we report that the virus extensively modulates the content of extracellular vesicles, enhancing viral dissemination. This underscores the critical interplay between the virus, host proteins, and extracellular vesicles.


Sujet(s)
Coronavirus humain OC43 , Vésicules extracellulaires , Protéomique , Virion , Virion/métabolisme , Humains , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/virologie , Coronavirus humain OC43/physiologie , Coronavirus humain OC43/métabolisme , Protéomique/méthodes , Protéome/métabolisme , Protéome/analyse , Exosomes/métabolisme , Exosomes/virologie , Infections à coronavirus/virologie , Infections à coronavirus/métabolisme , Lignée cellulaire , Interactions hôte-pathogène
5.
Viruses ; 16(7)2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39066286

RÉSUMÉ

The BK polyomavirus (BKPyV) is a small DNA non-enveloped virus whose infection is asymptomatic in most of the world's adult population. However, in cases of immunosuppression, the reactivation of the virus can cause various complications, and in particular, nephropathies in kidney transplant recipients or hemorrhagic cystitis in bone marrow transplant recipients. Recently, it was demonstrated that BKPyV virions can use extracellular vesicles to collectively traffic in and out of cells, thus exiting producing cells without cell lysis and entering target cells by diversified entry routes. By a comparison to other naked viruses, we investigated the possibility that BKPyV virions recruit the Endosomal-Sorting Complexes Required for Transport (ESCRT) machinery through late domains in order to hijack extracellular vesicles. We identified a single potential late domain in the BKPyV structural proteins, a YPX3L motif in the VP1 protein, and used pseudovirions to study the effect of point mutations found in a BKPyV clinical isolate or known to ablate the interaction of such a domain with the ESCRT machinery. Our results suggest that this domain is not involved in BKPyV association with extracellular vesicles but is crucial for capsomere interaction and thus viral particle assembly.


Sujet(s)
Motifs d'acides aminés , Virus BK , Protéines de capside , Vésicules extracellulaires , Virion , Assemblage viral , Virus BK/génétique , Virus BK/physiologie , Virus BK/métabolisme , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/virologie , Humains , Protéines de capside/métabolisme , Protéines de capside/génétique , Protéines de capside/composition chimique , Virion/métabolisme , Virion/génétique , Infections à polyomavirus/virologie , Infections à polyomavirus/métabolisme , Complexes de tri endosomique requis pour le transport/métabolisme , Complexes de tri endosomique requis pour le transport/génétique , Cellules HEK293
6.
Methods Mol Biol ; 2829: 237-246, 2024.
Article de Anglais | MEDLINE | ID: mdl-38951339

RÉSUMÉ

Virus-like particles (VLP) of the cowpea chlorotic mottle virus (CCMV), a plant virus, have been shown to be safe and noncytotoxic vehicles for delivering various cargos, including nucleic acids and peptides, and as scaffolds for presenting epitopes. Thus, CCMV-VLP have acquired increasing attention to be used in fields such as gene therapy, drug delivery, and vaccine development. Regardless of their production method, most reports purify CCMV-VLP through a series of ultracentrifugation steps using sucrose density gradient ultracentrifugation, which is a complex and time-consuming process. Here, the use of anion exchange chromatography is described as a one-step protocol for purification of CCMV-VLP produced by the insect cell-baculovirus expression vector system (IC-BEVS).


Sujet(s)
Bromovirus , Bromovirus/génétique , Animaux , Baculoviridae/génétique , Vecteurs génétiques/génétique , Chromatographie d'échange d'ions/méthodes , Virion/isolement et purification , Virion/génétique , Virion/métabolisme
7.
J Infect Dis ; 230(1): 78-85, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39052738

RÉSUMÉ

As use of human immunodeficiency virus (HIV) integrase strand transfer inhibitors (INSTI) increases and formulations are being developed for maintenance therapies and chemoprophylaxis, assessing virus suppression under INSTI-based regimens in prevention-relevant biologic compartments, such as the male genital tract, is timely. We used cell-source marker virion immunocapture to examine amplification of particle RNA then assessed the phylogenetic relatedness of seminal and blood viral sequences from men with HIV who were prescribed INSTI-based regimens. Seminal plasma immunocaptures yielded amplifiable virion RNA from 13 of 24 (54%) men, and the sequences were primarily associated with markers indicative of macrophage and resident dendritic cell sources. Genetic distances were greatest (>2%) between seminal virions and circulating proviruses, pointing to ongoing low-level expression from tissue-resident cells. While the low levels in semen predict an improbable likelihood of transmission, viruses with large genetic distances are expressed under potent INSTI therapy and have implications for determining epidemiologic linkages if adherence is suboptimal.


Sujet(s)
Infections à VIH , Inhibiteurs de l'intégrase du VIH , Sperme , Sperme/virologie , Humains , Mâle , Infections à VIH/traitement médicamenteux , Infections à VIH/virologie , Inhibiteurs de l'intégrase du VIH/pharmacologie , Inhibiteurs de l'intégrase du VIH/usage thérapeutique , Adulte , Phylogenèse , ARN viral/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Virion/métabolisme , Adulte d'âge moyen
8.
Nat Commun ; 15(1): 4906, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38851803

RÉSUMÉ

Various low-density lipoprotein receptors (LPRs) have been identified as entry factors for alphaviruses, and structures of the corresponding virion-receptor complexes have been determined. Here, we analyze the similarities and differences in the receptor binding modes of multiple alphaviruses to understand their ability to infect a wide range of hosts. We further discuss the challenges associated with the development of broad-spectrum treatment strategies against a diverse range of alphaviruses.


Sujet(s)
Alphavirus , Antiviraux , Récepteurs aux lipoprotéines LDL , Pénétration virale , Animaux , Humains , Alphavirus/effets des médicaments et des substances chimiques , Alphavirus/physiologie , Alphavirus/génétique , Infections à alphavirus/traitement médicamenteux , Infections à alphavirus/virologie , Antiviraux/usage thérapeutique , Antiviraux/pharmacologie , Liaison aux protéines , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs viraux/métabolisme , Récepteurs viraux/composition chimique , Virion/métabolisme , Pénétration virale/effets des médicaments et des substances chimiques
9.
Proc Natl Acad Sci U S A ; 121(24): e2400378121, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38830096

RÉSUMÉ

Epitranscriptomic RNA modifications have emerged as important regulators of the fate and function of viral RNAs. One prominent modification, the cytidine methylation 5-methylcytidine (m5C), is found on the RNA of HIV-1, where m5C enhances the translation of HIV-1 RNA. However, whether m5C functionally enhances the RNA of other pathogenic viruses remains elusive. Here, we surveyed a panel of commonly found RNA modifications on the RNA of hepatitis B virus (HBV) and found that HBV RNA is enriched with m5C as well as ten other modifications, at stoichiometries much higher than host messenger RNA (mRNA). Intriguingly, m5C is mostly found on the epsilon hairpin, an RNA element required for viral RNA encapsidation and reverse transcription, with these m5C mainly deposited by the cellular methyltransferase NSUN2. Loss of m5C from HBV RNA due to NSUN2 depletion resulted in a partial decrease in viral core protein (HBc) production, accompanied by a near-complete loss of the reverse transcribed viral DNA. Similarly, mutations introduced to remove the methylated cytidines resulted in a loss of HBc production and reverse transcription. Furthermore, pharmacological disruption of m5C deposition led to a significant decrease in HBV replication. Thus, our data indicate m5C methylations as a critical mediator of the epsilon elements' function in HBV virion production and reverse transcription, suggesting the therapeutic potential of targeting the m5C methyltransfer process on HBV epsilon as an antiviral strategy.


Sujet(s)
Cytidine , Virus de l'hépatite B , ARN viral , Transcription inverse , Virus de l'hépatite B/génétique , Virus de l'hépatite B/métabolisme , Virus de l'hépatite B/physiologie , ARN viral/génétique , ARN viral/métabolisme , Cytidine/analogues et dérivés , Cytidine/métabolisme , Cytidine/génétique , Humains , Transcription inverse/génétique , Méthylation , Réplication virale/génétique , Épigenèse génétique , Virion/métabolisme , Virion/génétique , Transcriptome
10.
Viruses ; 16(6)2024 Jun 09.
Article de Anglais | MEDLINE | ID: mdl-38932227

RÉSUMÉ

The HIV envelope glycoprotein (Env) is a trimeric protein that facilitates viral binding and fusion with target cells. As the sole viral protein on the HIV surface, Env is important both for immune responses to HIV and in vaccine designs. Targeting Env in clinical applications is challenging due to its heavy glycosylation, high genetic variability, conformational camouflage, and its low abundance on virions. Thus, there is a critical need to better understand this protein. Flow virometry (FV) is a useful methodology for phenotyping the virion surface in a high-throughput, single virion manner. To demonstrate the utility of FV to characterize Env, we stained HIV virions with a panel of 85 monoclonal antibodies targeting different regions of Env. A broad range of antibodies yielded robust staining of Env, with V3 antibodies showing the highest quantitative staining. A subset of antibodies tested in parallel on viruses produced in CD4+ T cell lines, HEK293T cells, and primary cells showed that the cellular model of virus production can impact Env detection. Finally, in addition to being able to highlight Env heterogeneity on virions, we show FV can sensitively detect differences in Env conformation when soluble CD4 is added to virions before staining.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Virion , Produits du gène env du virus de l'immunodéficience humaine , Humains , Produits du gène env du virus de l'immunodéficience humaine/composition chimique , Produits du gène env du virus de l'immunodéficience humaine/métabolisme , Produits du gène env du virus de l'immunodéficience humaine/génétique , Produits du gène env du virus de l'immunodéficience humaine/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Virion/métabolisme , Cellules HEK293 , Anticorps anti-VIH/immunologie , Anticorps monoclonaux/immunologie , Lymphocytes T CD4+/virologie , Lymphocytes T CD4+/immunologie , Infections à VIH/virologie
11.
Cell Host Microbe ; 32(7): 1050-1058.e7, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38870941

RÉSUMÉ

Viral genomes are most vulnerable to cellular defenses at the start of the infection. A family of jumbo phages related to phage ΦKZ, which infects Pseudomonas aeruginosa, assembles a protein-based phage nucleus to protect replicating phage DNA, but how it is protected prior to phage nucleus assembly is unclear. We find that host proteins related to membrane and lipid biology interact with injected phage protein, clustering in an early phage infection (EPI) vesicle. The injected virion RNA polymerase (vRNAP) executes early gene expression until phage genome separation from the vRNAP and the EPI vesicle, moving into the nascent proteinaceous phage nucleus. Enzymes involved in DNA replication and CRISPR/restriction immune nucleases are excluded by the EPI vesicle. We propose that the EPI vesicle is rapidly constructed with injected phage proteins, phage DNA, host lipids, and host membrane proteins to enable genome protection, early transcription, localized translation, and to ensure faithful genome transfer to the proteinaceous nucleus.


Sujet(s)
ADN viral , Génome viral , Phages de Pseudomonas , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virologie , Phages de Pseudomonas/génétique , Phages de Pseudomonas/métabolisme , ADN viral/génétique , Protéines virales/métabolisme , Protéines virales/génétique , Bactériophages/génétique , Bactériophages/physiologie , Virion/métabolisme , Réplication virale , DNA-directed RNA polymerases/métabolisme , DNA-directed RNA polymerases/génétique , Lipides , Réplication de l'ADN
12.
PLoS Pathog ; 20(6): e1012311, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38885273

RÉSUMÉ

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.


Sujet(s)
Virion , Zinc , Zinc/métabolisme , Virion/métabolisme , Protéines de capside/métabolisme , Assemblage viral/physiologie , Virus des plantes/métabolisme , Virus des plantes/physiologie , Maladies des plantes/virologie , Cystéine/métabolisme , Protéines virales/métabolisme , Morphogenèse
13.
J Biotechnol ; 391: 57-63, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-38851397

RÉSUMÉ

Antigen-presenting cells (APCs) play an important role in virus infection control by bridging innate and adaptive immune responses. Macrophages and dendritic cells (DCs) possess various surface receptors to recognize/internalize antigens, and antibody binding can enhance pathogen-opsonizing uptake by these APCs via interaction of antibody fragment crystallizable (Fc) domains with Fc receptors, evoking profound pathogen control in certain settings. Here, we examined phagocytosis-enhancing potential of Fc domains directly oriented on a retroviral virion/virus-like particle (VLP) surface. We generated an expression vector coding a murine Fc fragment fused to the transmembrane region (TM) of a retroviral envelope protein, deriving expression of the Fc-TM fusion protein on the transfected cell surface and production of virions incorporating the chimeric Fc upon co-transfection. Incubation of Fc-displaying simian immunodeficiency virus (SIV) with murine J774 macrophages and bone marrow-derived DCs derived Fc receptor-dependent enhanced uptake, being visualized by imaging cytometry. Alternative preparation of a murine leukemia virus (MLV) backbone-based Fc-displaying VLP loading an influenza virus hemagglutinin (HA) antigen resulted in enhanced HA internalization by macrophages, stating antigen compatibility of the design. Results show that the Fc-TM fusion molecule can be displayed on certain viruses/VLPs and may be utilized as a molecular adjuvant to facilitate APC antigen uptake.


Sujet(s)
Cellules présentatrices d'antigène , Cellules dendritiques , Fragments Fc des immunoglobulines , Virion , Animaux , Souris , Fragments Fc des immunoglobulines/génétique , Fragments Fc des immunoglobulines/métabolisme , Fragments Fc des immunoglobulines/immunologie , Cellules présentatrices d'antigène/immunologie , Cellules présentatrices d'antigène/métabolisme , Virion/métabolisme , Virion/génétique , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Macrophages/métabolisme , Macrophages/immunologie , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Protéines de fusion recombinantes/immunologie , Virus de l'immunodéficience simienne/immunologie , Virus de l'immunodéficience simienne/génétique , Glycoprotéine hémagglutinine du virus influenza/immunologie , Glycoprotéine hémagglutinine du virus influenza/génétique , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Lignée cellulaire , Virus de la leucémie murine/génétique , Phagocytose , Humains
14.
Int J Mol Sci ; 25(12)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38928103

RÉSUMÉ

The maturation of HIV-1 virions is a crucial process in viral replication. Although T-cells are a primary source of virus production, much of our understanding of virion maturation comes from studies using the HEK293T human embryonic kidney cell line. Notably, there is a lack of comparative analyses between T-cells and HEK293T cells in terms of virion maturation efficiency in existing literature. We previously developed an advanced virion visualization system based on the FRET principle, enabling the effective distinction between immature and mature virions via fluorescence microscopy. In this study, we utilized pseudotyped, single-round infectious viruses tagged with FRET labels (HIV-1 Gag-iFRET∆Env) derived from Jurkat (a human T-lymphocyte cell line) and HEK293T cells to evaluate their virion maturation rates. HEK293T-derived virions demonstrated a maturity rate of 81.79%, consistent with other studies and our previous findings. However, virions originating from Jurkat cells demonstrated a significantly reduced maturation rate of 68.67% (p < 0.0001). Correspondingly, viruses produced from Jurkat cells exhibited significantly reduced infectivity compared to those derived from HEK293T cells, with the relative infectivity measured at 65.3%. This finding is consistent with the observed relative maturation rate of viruses produced by Jurkat cells. These findings suggest that initiation of virion maturation directly correlates with viral infectivity. Our observation highlights the dynamic nature of virus-host interactions and their implications for virion production and infectivity.


Sujet(s)
Transfert d'énergie par résonance de fluorescence , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Virion , Humains , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/pathogénicité , Cellules HEK293 , Virion/métabolisme , Cellules Jurkat , Transfert d'énergie par résonance de fluorescence/méthodes , Réplication virale , Assemblage viral , Infections à VIH/virologie
15.
Viruses ; 16(6)2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38932164

RÉSUMÉ

The HIV-1 nucleocapsid protein (NC) is a multifunctional viral protein necessary for HIV-1 replication. Recent studies have demonstrated that reverse transcription (RT) completes in the intact viral capsid, and the timing of RT and uncoating are correlated. How the small viral core stably contains the ~10 kbp double stranded (ds) DNA product of RT, and the role of NC in this process, are not well understood. We showed previously that NC binds and saturates dsDNA in a non-specific electrostatic binding mode that triggers uniform DNA self-attraction, condensing dsDNA into a tight globule against extending forces up to 10 pN. In this study, we use optical tweezers and atomic force microscopy to characterize the role of NC's basic residues in dsDNA condensation. Basic residue mutations of NC lead to defective interaction with the dsDNA substrate, with the constant force plateau condensation observed with wild-type (WT) NC missing or diminished. These results suggest that NC's high positive charge is essential to its dsDNA condensing activity, and electrostatic interactions involving NC's basic residues are responsible in large part for the conformation, size, and stability of the dsDNA-protein complex inside the viral core. We observe DNA re-solubilization and charge reversal in the presence of excess NC, consistent with the electrostatic nature of NC-induced DNA condensation. Previous studies of HIV-1 replication in the presence of the same cationic residue mutations in NC showed significant defects in both single- and multiple-round viral infectivity. Although NC participates in many stages of viral replication, our results are consistent with the hypothesis that cationic residue mutations inhibit genomic DNA condensation, resulting in increased premature capsid uncoating and contributing to viral replication defects.


Sujet(s)
ADN viral , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Transcription inverse , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/composition chimique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , ADN viral/génétique , ADN viral/métabolisme , Produits du gène gag du virus de l'immunodéficience humaine/métabolisme , Produits du gène gag du virus de l'immunodéficience humaine/génétique , Produits du gène gag du virus de l'immunodéficience humaine/composition chimique , Humains , Cations/métabolisme , Réplication virale , Microscopie à force atomique , Virion/métabolisme , Virion/génétique , Virion/composition chimique , Mutation
16.
Nature ; 630(8017): 712-719, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38839957

RÉSUMÉ

Genetic screens have transformed our ability to interrogate cellular factor requirements for viral infections1,2, but most current approaches are limited in their sensitivity, biased towards early stages of infection and provide only simplistic phenotypic information that is often based on survival of infected cells2-4. Here, by engineering human cytomegalovirus to express single guide RNA libraries directly from the viral genome, we developed virus-encoded CRISPR-based direct readout screening (VECOS), a sensitive, versatile, viral-centric approach that enables profiling of different stages of viral infection in a pooled format. Using this approach, we identified hundreds of host dependency and restriction factors and quantified their direct effects on viral genome replication, viral particle secretion and infectiousness of secreted particles, providing a multi-dimensional perspective on virus-host interactions. These high-resolution measurements reveal that perturbations altering late stages in the life cycle of human cytomegalovirus (HCMV) mostly regulate viral particle quality rather than quantity, establishing correct virion assembly as a critical stage that is heavily reliant on virus-host interactions. Overall, VECOS facilitates systematic high-resolution dissection of the role of human proteins during the infection cycle, providing a roadmap for in-depth study of host-herpesvirus interactions.


Sujet(s)
Systèmes CRISPR-Cas , Infections à cytomégalovirus , Cytomegalovirus , Interactions hôte-pathogène , , Réplication virale , Humains , Lignée cellulaire , Systèmes CRISPR-Cas/génétique , Cytomegalovirus/génétique , Cytomegalovirus/physiologie , Infections à cytomégalovirus/génétique , Infections à cytomégalovirus/virologie , Génome viral/génétique , Interactions hôte-pathogène/génétique , /génétique , /métabolisme , Virion/génétique , Virion/métabolisme , Assemblage viral/génétique , Libération de particules virales/génétique , Réplication virale/génétique
17.
Viruses ; 16(6)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38932209

RÉSUMÉ

A proteomics analysis of purified rabies virus (RABV) revealed 47 entrapped host proteins within the viral particles. Out of these, 11 proteins were highly disordered. Our study was particularly focused on five of the RABV-entrapped mouse proteins with the highest levels of disorder: Neuromodulin, Chmp4b, DnaJB6, Vps37B, and Wasl. We extensively utilized bioinformatics tools, such as FuzDrop, D2P2, UniProt, RIDAO, STRING, AlphaFold, and ELM, for a comprehensive analysis of the intrinsic disorder propensity of these proteins. Our analysis suggested that these disordered host proteins might play a significant role in facilitating the rabies virus pathogenicity, immune system evasion, and the development of antiviral drug resistance. Our study highlighted the complex interaction of the virus with its host, with a focus on how the intrinsic disorder can play a crucial role in virus pathogenic processes, and suggested that these intrinsically disordered proteins (IDPs) and disorder-related host interactions can also be a potential target for therapeutic strategies.


Sujet(s)
Protéines intrinsèquement désordonnées , Virus de la rage , Virion , Virus de la rage/physiologie , Animaux , Souris , Protéines intrinsèquement désordonnées/métabolisme , Protéines intrinsèquement désordonnées/composition chimique , Virion/métabolisme , Protéomique , Interactions hôte-pathogène , Rage (maladie)/virologie , Biologie informatique/méthodes , Protéines virales/métabolisme , Protéines virales/génétique , Protéines virales/composition chimique
18.
J Mol Biol ; 436(16): 168639, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38838849

RÉSUMÉ

HIV-1 Gag polyprotein plays a pivotal role in assembly and budding of new particles, by specifically packaging two copies of viral gRNA in the host cell cytoplasm and selecting the cell plasma membrane for budding. Both gRNA and membrane selections are thought to be mediated by the compact form of Gag. This compact form binds to gRNA through both its matrix (MA) and nucleocapsid (NC) domains in the cytoplasm. At the plasma membrane, the membrane competes with gRNA for Gag binding, resulting in a transition to the extended form of Gag found in immature particles with MA bound to membrane lipids and NC to gRNA. The Gag compact form was previously evidenced in vitro. Here, we demonstrated the compact form of Gag in cells by confocal microscopy, using a bimolecular fluorescence complementation approach with a split-GFP bipartite system. Using wild-type Gag and Gag mutants, we showed that the compact form is highly dependent on the binding of MA and NC domains to RNA, as well as on interactions between MA and CA domains. In contrast, Gag multimerization appears to be less critical for the accumulation of the compact form. Finally, mutations altering the formation of Gag compact form led to a strong reduction in viral particle production and infectivity, revealing its key role in the production of infectious viral particles.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Assemblage viral , Produits du gène gag du virus de l'immunodéficience humaine , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , Produits du gène gag du virus de l'immunodéficience humaine/métabolisme , Produits du gène gag du virus de l'immunodéficience humaine/génétique , Produits du gène gag du virus de l'immunodéficience humaine/composition chimique , Assemblage viral/génétique , Humains , Virion/métabolisme , Virion/génétique , Liaison aux protéines , ARN viral/métabolisme , ARN viral/génétique , Membrane cellulaire/métabolisme , Membrane cellulaire/virologie
19.
Nat Commun ; 15(1): 4542, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38806525

RÉSUMÉ

The Crimean-Congo hemorrhagic fever virus (CCHFV) is an emerging pathogen of the Orthonairovirus genus that can cause severe and often lethal hemorrhagic diseases in humans. CCHFV has a broad tropism and can infect a variety of species and tissues. Here, by using gene silencing, blocking antibodies or soluble receptor fragments, we identify the low-density lipoprotein receptor (LDL-R) as a CCHFV entry factor. The LDL-R facilitates binding of CCHFV particles but does not allow entry of Hazara virus (HAZV), another member of the genus. In addition, we show that apolipoprotein E (apoE), an exchangeable protein that mediates LDL/LDL-R interaction, is incorporated on CCHFV particles, though not on HAZV particles, and enhances their specific infectivity by promoting an LDL-R dependent entry. Finally, we show that molecules that decrease LDL-R from the surface of target cells could inhibit CCHFV infection. Our study highlights that CCHFV takes advantage of a lipoprotein receptor and recruits its natural ligand to promote entry into cells.


Sujet(s)
Apolipoprotéines E , Virus de la fièvre hémorragique de Crimée-Congo , Récepteurs aux lipoprotéines LDL , Pénétration virale , Humains , Récepteurs aux lipoprotéines LDL/métabolisme , Apolipoprotéines E/métabolisme , Apolipoprotéines E/génétique , Virus de la fièvre hémorragique de Crimée-Congo/métabolisme , Virus de la fièvre hémorragique de Crimée-Congo/physiologie , Animaux , Cellules HEK293 , Chlorocebus aethiops , Fièvre hémorragique de Crimée-Congo/virologie , Fièvre hémorragique de Crimée-Congo/métabolisme , Virion/métabolisme , Cellules Vero
20.
Biomolecules ; 14(5)2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38785977

RÉSUMÉ

Host restriction factor SERINC5 (SER5) incorporates into the HIV-1 membrane and inhibits infectivity by a poorly understood mechanism. Recently, SER5 was found to exhibit scramblase-like activity leading to the externalization of phosphatidylserine (PS) on the viral surface, which has been proposed to be responsible for SER5's antiviral activity. This and other reports that document modulation of HIV-1 infectivity by viral lipid composition prompted us to investigate the role of PS in regulating SER5-mediated HIV-1 restriction. First, we show that the level of SER5 incorporation into virions correlates with an increase in PS levels in the outer leaflet of the viral membrane. We developed an assay to estimate the PS distribution across the viral membrane and found that SER5, but not SER2, which lacks antiviral activity, abrogates PS asymmetry by externalizing this lipid. Second, SER5 incorporation diminished the infectivity of pseudoviruses produced from cells lacking a flippase subunit CDC50a and, therefore, exhibited a higher baseline level of surface-accessible PS. Finally, exogenous manipulation of the viral PS levels utilizing methyl-alpha-cyclodextrin revealed a lack of correlation between external PS and virion infectivity. Taken together, our study implies that the increased PS exposure to SER5-containing virions itself is not directly linked to HIV-1 restriction.


Sujet(s)
VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Protéines membranaires , Phosphatidylsérine , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Phosphatidylsérine/métabolisme , Humains , Protéines membranaires/métabolisme , Virion/métabolisme , Cellules HEK293 , Membrane cellulaire/métabolisme , Infections à VIH/virologie , Infections à VIH/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE