Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.496
Filter
1.
Nat Commun ; 15(1): 5514, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951492

ABSTRACT

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.


Subject(s)
HIV-1 , Immunity, Innate , Macrophages , Proto-Oncogene Proteins , Trans-Activators , vpr Gene Products, Human Immunodeficiency Virus , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , vpr Gene Products, Human Immunodeficiency Virus/metabolism , vpr Gene Products, Human Immunodeficiency Virus/genetics , HIV-1/physiology , HIV-1/immunology , Trans-Activators/metabolism , Trans-Activators/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , HIV Infections/immunology , HIV Infections/virology , HIV Infections/genetics , HEK293 Cells , Virion/metabolism , Protein Serine-Threonine Kinases
2.
Methods Mol Biol ; 2829: 237-246, 2024.
Article in English | MEDLINE | ID: mdl-38951339

ABSTRACT

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).


Subject(s)
Bromovirus , Bromovirus/genetics , Animals , Baculoviridae/genetics , Genetic Vectors/genetics , Chromatography, Ion Exchange/methods , Virion/isolation & purification , Virion/genetics , Virion/metabolism
3.
PLoS Pathog ; 20(6): e1012311, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38885273

ABSTRACT

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


Subject(s)
Virion , Zinc , Zinc/metabolism , Virion/metabolism , Capsid Proteins/metabolism , Virus Assembly/physiology , Plant Viruses/metabolism , Plant Viruses/physiology , Plant Diseases/virology , Cysteine/metabolism , Viral Proteins/metabolism , Morphogenesis
4.
J Biotechnol ; 391: 57-63, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38851397

ABSTRACT

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.


Subject(s)
Antigen-Presenting Cells , Dendritic Cells , Immunoglobulin Fc Fragments , Virion , Animals , Mice , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Fc Fragments/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Virion/metabolism , Virion/genetics , Dendritic Cells/immunology , Dendritic Cells/metabolism , Macrophages/metabolism , Macrophages/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/immunology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Cell Line , Leukemia Virus, Murine/genetics , Phagocytosis , Humans
5.
Viruses ; 16(6)2024 May 29.
Article in English | MEDLINE | ID: mdl-38932164

ABSTRACT

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.


Subject(s)
DNA, Viral , HIV-1 , Reverse Transcription , HIV-1/genetics , HIV-1/physiology , HIV-1/chemistry , HIV-1/metabolism , DNA, Viral/genetics , DNA, Viral/metabolism , gag Gene Products, Human Immunodeficiency Virus/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/chemistry , Humans , Cations/metabolism , Virus Replication , Microscopy, Atomic Force , Virion/metabolism , Virion/genetics , Virion/chemistry , Mutation
6.
Int J Mol Sci ; 25(12)2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38928103

ABSTRACT

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.


Subject(s)
Fluorescence Resonance Energy Transfer , HIV-1 , Virion , Humans , HIV-1/physiology , HIV-1/pathogenicity , HEK293 Cells , Virion/metabolism , Jurkat Cells , Fluorescence Resonance Energy Transfer/methods , Virus Replication , Virus Assembly , HIV Infections/virology
7.
Nature ; 630(8017): 712-719, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839957

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems , Cytomegalovirus Infections , Cytomegalovirus , Host-Pathogen Interactions , RNA, Guide, CRISPR-Cas Systems , Virus Replication , Humans , Cell Line , CRISPR-Cas Systems/genetics , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Genome, Viral/genetics , Host-Pathogen Interactions/genetics , RNA, Guide, CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/metabolism , Virion/genetics , Virion/metabolism , Virus Assembly/genetics , Virus Release/genetics , Virus Replication/genetics
8.
Nat Commun ; 15(1): 4906, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851803

ABSTRACT

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.


Subject(s)
Alphavirus , Antiviral Agents , Receptors, LDL , Virus Internalization , Animals , Humans , Alphavirus/drug effects , Alphavirus/physiology , Alphavirus/genetics , Alphavirus Infections/drug therapy , Alphavirus Infections/virology , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Protein Binding , Receptors, LDL/metabolism , Receptors, LDL/genetics , Receptors, Virus/metabolism , Receptors, Virus/chemistry , Virion/metabolism , Virus Internalization/drug effects
9.
Proc Natl Acad Sci U S A ; 121(24): e2400378121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38830096

ABSTRACT

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.


Subject(s)
Cytidine , Hepatitis B virus , RNA, Viral , Reverse Transcription , Hepatitis B virus/genetics , Hepatitis B virus/metabolism , Hepatitis B virus/physiology , RNA, Viral/genetics , RNA, Viral/metabolism , Cytidine/analogs & derivatives , Cytidine/metabolism , Cytidine/genetics , Humans , Reverse Transcription/genetics , Methylation , Virus Replication/genetics , Epigenesis, Genetic , Virion/metabolism , Virion/genetics , Transcriptome
10.
Viruses ; 16(6)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38932209

ABSTRACT

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.


Subject(s)
Intrinsically Disordered Proteins , Rabies virus , Virion , Rabies virus/physiology , Animals , Mice , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/chemistry , Virion/metabolism , Proteomics , Host-Pathogen Interactions , Rabies/virology , Computational Biology/methods , Viral Proteins/metabolism , Viral Proteins/genetics , Viral Proteins/chemistry
11.
Viruses ; 16(6)2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38932227

ABSTRACT

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.


Subject(s)
HIV-1 , Virion , env Gene Products, Human Immunodeficiency Virus , Humans , env Gene Products, Human Immunodeficiency Virus/chemistry , env Gene Products, Human Immunodeficiency Virus/metabolism , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology , HIV-1/genetics , HIV-1/physiology , HIV-1/immunology , Virion/metabolism , HEK293 Cells , HIV Antibodies/immunology , Antibodies, Monoclonal/immunology , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/virology
12.
Virology ; 596: 110120, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38805801

ABSTRACT

Glycoprotein C (gC), one of ∼12 HSV-1 envelope glycoproteins, carries out several important functions during infection, including the enhancement of virion attachment by binding to host cell heparan sulfate proteoglycans (HSPG). Here we report that gC can also enhance the release of cell-free progeny virions at the end of the infectious cycle. This activity was observed in multiple cellular contexts including Vero cells and immortalized human keratinocytes. In the absence of gC, progeny virions bound more tightly to infected cells, suggesting that gC promotes the detachment of virions from the infected cell surface. Given this finding, we analyzed the biochemical interactions that tether progeny virions to cells and report evidence for two distinct modes of binding. One is consistent with a direct interaction between gC and HSPG, whereas the other is gC-independent and likely does not involve HSPG. Together, our results i) identify a novel function for a long-studied HSV-1 glycoprotein, and ii) demonstrate that the extracellular release of HSV-1 virions is a dynamic process involving multiple viral and host components.


Subject(s)
Herpesvirus 1, Human , Viral Envelope Proteins , Virion , Virus Release , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/metabolism , Humans , Chlorocebus aethiops , Vero Cells , Animals , Virion/metabolism , Heparan Sulfate Proteoglycans/metabolism , Keratinocytes/virology , Keratinocytes/metabolism
13.
J Virol ; 98(6): e0023524, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38775478

ABSTRACT

Baculoviruses enter insect midgut epithelial cells via a set of occlusion-derived virion (ODV) envelope proteins called per os infectivity factors (PIFs). P74 of Autographa californica multicapsid nucleopolyhedrovirus (AcMNPV), which was the first identified PIF, is cleaved by an endogenous proteinase embedded within the occlusion body during per os infection, but the target site(s) and function of the cleavage have not yet been ascertained. Here, based on bioinformatics analyses, we report that cleavage was predicted at an arginine and lysine-rich region in the middle of P74. A series of recombinant viruses with site-directed mutants in this region of P74 were generated. R325 or R334 was identified as primary cleavage site. In addition, we showed that P74 is also cleaved by brush border membrane vesicles (BBMV) of the host insect at R325 or R334, instead of R195, R196, and R199, as previously reported. Simultaneous mutations in R195, R196, and R199 lead to instability of P74 during ODV release. Bioassays showed that mutations at both R325 and R334 significantly affected oral infectivity. Taken together, our data show that both R325 and R334 of AcMNPV P74 are the primary cleavage site for both occlusion body endogenous proteinase and BBMV proteinase during ODV release and are critical for oral infection. IMPORTANCE: Cleavage of viral envelope proteins is usually an important trigger for viral entry into host cells. Baculoviruses are insect-specific viruses that infect host insects via the oral route. P74, a per os infectivity factor of baculoviruses, is cleaved during viral entry. However, the function and precise cleavage sites of P74 remain unknown. In this study, we found that R325 or R334 between the N- and C-conserved domains of P74 was the primary cleavage site by proteinase either from the occlusion body or host midgut. The biological significance of cleavage seems to be the release of the potential fusion peptide at the N-terminus of the cleaved C-terminal P74. Our results shed light on the cleavage model of P74 and imply its role in membrane fusion in baculovirus per os infection.


Subject(s)
Nucleopolyhedroviruses , Animals , Nucleopolyhedroviruses/metabolism , Nucleopolyhedroviruses/genetics , Peptide Hydrolases/metabolism , Peptide Hydrolases/genetics , Virus Internalization , Sf9 Cells , Spodoptera , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Microvilli/metabolism , Microvilli/virology , Virion/metabolism , Occlusion Bodies, Viral/metabolism
14.
Emerg Microbes Infect ; 13(1): 2356149, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38747061

ABSTRACT

Lassa virus (LASV), a risk-group 4 pathogen, must be handled in biosafety level-4 (BSL-4) conditions, thereby limiting its research and antiviral development. Here, we developed a novel LASV reverse genetics system which, to our knowledge, is the first to study the complete LASV life cycle under BSL-2 conditions. Viral particles can be produced efficiently when LASV minigenomic RNA harbouring minimal viral cis-elements and reporter genes is transfected into a helper cell line stably expressing viral NP, GP, Z and L proteins. The resulting defective virions, named LASVmg, can propagate only in the helper cell line, providing a BSL-2 model to study the complete LASV life cycle. Using this model, we found that a previously reported cellular receptor α-dystroglycan is dispensable for LASVmg infection. Furthermore, we showed that ribavirin can inhibit LASVmg infection by inducing viral mutations. This new BSL-2 system should facilitate studying the LASV life cycle and screening antivirals.


Subject(s)
Lassa virus , Reverse Genetics , Lassa virus/genetics , Lassa virus/physiology , Reverse Genetics/methods , Humans , Animals , Antiviral Agents/pharmacology , Chlorocebus aethiops , Cell Line , Virus Replication , Lassa Fever/virology , Ribavirin/pharmacology , Vero Cells , Containment of Biohazards , Genome, Viral , Virion/genetics , Virion/metabolism
15.
Nat Microbiol ; 9(6): 1417-1426, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38783022

ABSTRACT

Ebola virus and other orthoebolaviruses cause severe haemorrhagic fevers in humans, with very high case fatality rates. Their non-segmented single-stranded RNA genome encodes only seven structural proteins and a small number of non-structural proteins to facilitate the virus life cycle. The basics of this life cycle are well established, but recent advances have substantially increased our understanding of its molecular details, including the viral and host factors involved. Here we provide a comprehensive overview of our current knowledge of the molecular details of the orthoebolavirus life cycle, with a special focus on proviral host factors. We discuss the multistep entry process, viral RNA synthesis in specialized phase-separated intracellular compartments called inclusion bodies, the expression of viral proteins and ultimately the assembly of new virus particles and their release at the cell surface. In doing so, we integrate recent studies into the increasingly detailed model that has developed for these fundamental aspects of orthoebolavirus biology.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , RNA, Viral , Ebolavirus/genetics , Ebolavirus/physiology , Humans , Hemorrhagic Fever, Ebola/virology , RNA, Viral/metabolism , RNA, Viral/genetics , Virus Replication , Viral Proteins/metabolism , Viral Proteins/genetics , Virus Assembly , Virus Internalization , Genome, Viral , Animals , Virion/metabolism , Virion/genetics , Host-Pathogen Interactions
16.
Nat Commun ; 15(1): 4542, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806525

ABSTRACT

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.


Subject(s)
Apolipoproteins E , Hemorrhagic Fever Virus, Crimean-Congo , Receptors, LDL , Virus Internalization , Humans , Receptors, LDL/metabolism , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Hemorrhagic Fever Virus, Crimean-Congo/metabolism , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Animals , HEK293 Cells , Chlorocebus aethiops , Hemorrhagic Fever, Crimean/virology , Hemorrhagic Fever, Crimean/metabolism , Virion/metabolism , Vero Cells
17.
Protein Expr Purif ; 220: 106502, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38754753

ABSTRACT

Adeno-associated Virus (AAV) is a promising vector for gene therapy. However, few studies have focused on producing virus-like particles (VLPs) of AAV in cells, especially in E. coli. In this study, we describe a method to produce empty VP3-only VLPs of AAV2 in E. coli by co-expressing VP3 and assembly-activating protein (AAP) of AAV2. Although the yields of VLPs produced with our method were low, the VLPs were able to self-assemble in E. coli without the need of in vitro capsid assembly. The produced VLPs were characterized by immunological detection and transmission electron microscopy (TEM). In conclusion, this study demonstrated that capsid assembly of AAV2 is possible in E. coli, and E. coli may be a candidate system for production of VLPs of AAV.


Subject(s)
Capsid Proteins , Dependovirus , Escherichia coli , Escherichia coli/genetics , Escherichia coli/metabolism , Dependovirus/genetics , Capsid Proteins/genetics , Capsid Proteins/metabolism , Capsid Proteins/biosynthesis , Virion/genetics , Virion/metabolism , Virus Assembly , Genetic Vectors/metabolism , Genetic Vectors/genetics , Genetic Vectors/chemistry , Parvovirinae/genetics , Humans
18.
Commun Biol ; 7(1): 590, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755280

ABSTRACT

Infection of bacteria by phages is a complex multi-step process that includes specific recognition of the host cell, creation of a temporary breach in the host envelope, and ejection of viral DNA into the bacterial cytoplasm. These steps must be perfectly regulated to ensure efficient infection. Here we report the dual function of the tail completion protein gp16.1 of bacteriophage SPP1. First, gp16.1 has an auxiliary role in assembly of the tail interface that binds to the capsid connector. Second, gp16.1 is necessary to ensure correct routing of phage DNA to the bacterial cytoplasm. Viral particles assembled without gp16.1 are indistinguishable from wild-type virions and eject DNA normally in vitro. However, they release their DNA to the extracellular space upon interaction with the host bacterium. The study shows that a highly conserved tail completion protein has distinct functions at two essential steps of the virus life cycle in long-tailed phages.


Subject(s)
Viral Tail Proteins , Viral Tail Proteins/metabolism , Viral Tail Proteins/genetics , Bacteriophages/genetics , Bacteriophages/physiology , Bacteriophages/metabolism , DNA, Viral/metabolism , DNA, Viral/genetics , Virion/metabolism
19.
Retrovirology ; 21(1): 10, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38778414

ABSTRACT

BACKGROUND: Detection of viruses by host pattern recognition receptors induces the expression of type I interferon (IFN) and IFN-stimulated genes (ISGs), which suppress viral replication. Numerous studies have described HIV-1 as a poor activator of innate immunity in vitro. The exact role that the viral capsid plays in this immune evasion is not fully understood. RESULTS: To better understand the role of the HIV-1 capsid in sensing we tested the effect of making HIV-1 by co-expressing a truncated Gag that encodes the first 107 amino acids of capsid fused with luciferase or GFP, alongside wild type Gag-pol. We found that unlike wild type HIV-1, viral particles produced with a mixture of wild type and truncated Gag fused to luciferase or GFP induced a potent IFN response in THP-1 cells and macrophages. Innate immune activation by Gag-fusion HIV-1 was dependent on reverse transcription and DNA sensor cGAS, suggesting activation of an IFN response by viral DNA. Further investigation revealed incorporation of the Gag-luciferase/GFP fusion proteins into viral particles that correlated with subtle defects in wild type Gag cleavage and a diminished capacity to saturate restriction factor TRIM5α, likely due to aberrant particle formation. We propose that expression of the Gag fusion protein disturbs the correct cleavage and maturation of wild type Gag, yielding viral particles that are unable to effectively shield viral DNA from detection by innate sensors including cGAS. CONCLUSIONS: These data highlight the crucial role of capsid in innate evasion and support growing literature that disruption of Gag cleavage and capsid formation induces a viral DNA- and cGAS-dependent innate immune response. Together these data demonstrate a protective role for capsid and suggest that antiviral activity of capsid-targeting antivirals may benefit from enhanced innate and adaptive immunity in vivo.


Subject(s)
HIV-1 , Immunity, Innate , Nucleotidyltransferases , gag Gene Products, Human Immunodeficiency Virus , HIV-1/immunology , HIV-1/genetics , HIV-1/physiology , Humans , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology , gag Gene Products, Human Immunodeficiency Virus/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Antiviral Restriction Factors , Macrophages/immunology , Macrophages/virology , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , THP-1 Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/immunology , Immune Evasion , Capsid/metabolism , Capsid/immunology , Virus Replication , Virion/metabolism , Virion/genetics , Virion/immunology , Host-Pathogen Interactions/immunology , DNA, Viral/genetics , Cell Line
20.
Commun Biol ; 7(1): 634, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796621

ABSTRACT

Ebola virus (EBOV) matrix protein VP40 can assemble and bud as virus-like particles (VLPs) when expressed alone in mammalian cells. Nucleoprotein (NP) could be recruited to VLPs as inclusion body (IB) when co-expressed, and increase VLP production. However, the mechanism behind it remains unclear. Here, we use a computational approach to study NP-VP40 interactions. Our simulations indicate that NP may enhance VLP production through stabilizing VP40 filaments and accelerating the VLP budding step. Further, both the relative timing and amount of NP expression compared to VP40 are important for the effective production of IB-containing VLPs. We predict that relative NP/VP40 expression ratio and time are important for efficient production of IB-containing VLPs. We conclude that disrupting the expression timing and amount of NP and VP40 could provide new avenues to treat EBOV infection. This work provides quantitative insights into EBOV proteins interactions and how virion generation and drug efficacy could be influenced.


Subject(s)
Ebolavirus , Viral Core Proteins , Ebolavirus/metabolism , Viral Core Proteins/metabolism , Viral Core Proteins/genetics , Humans , Virion/metabolism , Virion/genetics , Nucleoproteins/metabolism , Nucleoproteins/genetics , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/genetics , Hemorrhagic Fever, Ebola/virology , Hemorrhagic Fever, Ebola/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...