Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 14.261
Filter
1.
Methods Mol Biol ; 2848: 249-257, 2025.
Article in English | MEDLINE | ID: mdl-39240527

ABSTRACT

The production of Adeno-associated virus (AAV) vectors in the lab setting has typically involved expression in adherent cells followed by purification through ultracentrifugation in density gradients. This production method is, however, not easily scalable, presents high levels of cellular impurities that co-purify with the virus, and results in a mixture of empty and full capsids. Here we describe a detailed AAV production protocol that overcomes these limitations through AAV expression in suspension cells followed by AAV affinity purification and AAV polishing to separate empty and full capsids, resulting in high yields of ultra-pure AAV that is highly enriched in full capsids.


Subject(s)
Dependovirus , Genetic Vectors , Dependovirus/genetics , Dependovirus/isolation & purification , Genetic Vectors/genetics , Humans , Capsid/chemistry , Capsid/metabolism , Virion/isolation & purification , Virion/genetics , HEK293 Cells , Chromatography, Affinity/methods , Ultracentrifugation/methods , Capsid Proteins/isolation & purification , Capsid Proteins/genetics , Capsid Proteins/chemistry , Capsid Proteins/metabolism
2.
Viruses ; 16(9)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39339924

ABSTRACT

Rabies virus (RABV) is among the first recognized viruses of public health concern and has historically contributed to the development of viral vaccines. Despite these significances, the three-dimensional structure of the RABV virion remains unknown due to the challenges in isolating structurally homogenous virion samples in sufficient quantities needed for structural investigation. Here, by combining the capabilities of cryogenic electron tomography (cryoET) and microscopy (cryoEM), we determined the three-dimensional structure of the wild-type RABV virion. Tomograms of RABV virions reveal a high level of structural heterogeneity among the bullet-shaped virion particles encompassing the glycoprotein (G) trimer-decorated envelope and the nucleocapsid composed of RNA, nucleoprotein (N), and matrix protein (M). The structure of the trunk region of the virion was determined by cryoEM helical reconstruction, revealing a one-start N-RNA helix bound by a single layer of M proteins at an N:M ratio of 1. The N-M interaction differs from that in fellow rhabdovirus vesicular stomatitis virus (VSV), which features two layers of M stabilizing the N-RNA helix at an M:N ratio of 2. These differences in both M-N stoichiometry and binding allow RABV to flex its N-RNA helix more freely and point to different mechanisms of viral assembly between these two bullet-shaped rhabdoviruses.


Subject(s)
Cryoelectron Microscopy , Rabies virus , Virion , Rabies virus/ultrastructure , Rabies virus/chemistry , Virion/ultrastructure , Animals , RNA, Viral/genetics , RNA, Viral/metabolism , Electron Microscope Tomography , Models, Molecular , Nucleocapsid/ultrastructure , Nucleocapsid/metabolism , Nucleocapsid/chemistry , Rabies/virology , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism , Viral Matrix Proteins/ultrastructure , Viral Matrix Proteins/genetics
3.
Viruses ; 16(9)2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39339982

ABSTRACT

Our examination of RNA helicases for effects on HIV-1 protein production and particle assembly identified Rocaglamide (RocA), a known modulator of eIF4A1 function, as an inhibitor of HIV-1 replication in primary CD4+ T cells and three cell systems. HIV-1 attenuation by low-nM RocA doses was associated with reduced viral particle formation without a marked decrease in Gag production. Rather, the co-localization of Gag and HIV-1 genomic RNA (gRNA) assemblies was impaired by RocA treatment in a reversible fashion. Ribonucleoprotein (RNP) immunoprecipitation studies recapitulated the loss of Gag-gRNA assemblies upon RocA treatment. Parallel biophysical studies determined that neither RocA nor eIF4A1 independently affected the ability of Gag to interact with viral RNA, but together, they distorted the structure of the HIV-1 RNP visualized by electron microscopy. Taken together, several lines of evidence indicate that RocA induces stable binding of eIF4A1 onto the viral RNA genome in a manner that interferes with the ordered assembly of Gag along Gag-gRNA assemblies required to generate infectious virions.


Subject(s)
Benzofurans , HIV-1 , RNA, Viral , Virus Replication , gag Gene Products, Human Immunodeficiency Virus , HIV-1/drug effects , HIV-1/physiology , HIV-1/genetics , Humans , Virus Replication/drug effects , RNA, Viral/metabolism , RNA, Viral/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism , gag Gene Products, Human Immunodeficiency Virus/genetics , Benzofurans/pharmacology , Virion/drug effects , Virion/metabolism , CD4-Positive T-Lymphocytes/virology , Virus Assembly/drug effects , HIV Infections/virology , HIV Infections/drug therapy , Eukaryotic Initiation Factor-4A/metabolism , Genome, Viral/drug effects , Protein Binding , Cell Line
4.
Phytomedicine ; 134: 156015, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39244942

ABSTRACT

BACKGROUND: Pseudorabies virus (PRV), a member of the family Herpesviridae, is responsible for significant economic losses in the pig industry and has recently been associated with human viral encephalitis, leading to severe neurological symptoms post-recovery. Despite the widespread impact of PRV, there are currently no approved effective drugs for treating PRV-related diseases in humans or pigs. Therefore, the exploration and discovery of safe and effective drugs for the prevention and treatment of PRV infection is of paramount importance. PURPOSE: The objective of this study is to screen and identify natural compounds with antiviral activity against PRV. METHODS: First, we used a strain of PRV with green fluorescent protein (PRV-GFP) to screen a natural product chemical library to identify potential antiviral drugs. Next, we assessed the antiviral abilities of salvianolic acid A (SAA) in vitro using virus titer assay, qPCR, and IFA. We investigated the mechanisms of SAA's antiviral activity through viral attachment, internalization, inactivation, and nuclease digestion assay. Finally, we evaluated the efficacy of SAA in inactivating PRV using mice as the experimental subjects. RESULTS: This study screened 206 natural compounds for anti-PRV activity in vitro, resulting in the identification of seven potential antiviral agents. Notably, SAA emerged as a promising candidate with significant anti-PRV activity. The mechanism of action may be that SAA can directly inactivate the virus by disrupting viral envelope. In vivo experiments have shown that pre-incubation of SAA and PRV can effectively inhibit the infectivity and pathogenicity of PRV in mice. CONCLUSION: This study offers valuable insights into the antiviral properties of SAA, potentially informing strategies for controlling PRV epidemics and treating related diseases in both humans and animals.


Subject(s)
Antiviral Agents , Herpesvirus 1, Suid , Pseudorabies , Herpesvirus 1, Suid/drug effects , Animals , Antiviral Agents/pharmacology , Pseudorabies/drug therapy , Pseudorabies/virology , Mice , Caffeic Acids/pharmacology , Lactates/pharmacology , Virus Internalization/drug effects , Virion/drug effects , Swine , Virus Attachment/drug effects , Cell Line , Female
5.
PLoS Pathog ; 20(9): e1011810, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39226318

ABSTRACT

The viral capsid performs critical functions during HIV-1 infection and is a validated target for antiviral therapy. Previous studies have established that the proper structure and stability of the capsid are required for efficient HIV-1 reverse transcription in target cells. Moreover, it has recently been demonstrated that permeabilized virions and purified HIV-1 cores undergo efficient reverse transcription in vitro when the capsid is stabilized by addition of the host cell metabolite inositol hexakisphosphate (IP6). However, the molecular mechanism by which the capsid promotes reverse transcription is undefined. Here we show that wild type HIV-1 virions can undergo efficient reverse transcription in vitro in the absence of a membrane-permeabilizing agent. This activity, originally termed "natural endogenous reverse transcription" (NERT), depends on expression of the viral envelope glycoprotein during virus assembly and its incorporation into virions. Truncation of the gp41 cytoplasmic tail markedly reduced NERT activity, suggesting that gp41 licenses the entry of nucleotides into virions. By contrast to reverse transcription in permeabilized virions, NERT required neither the addition of IP6 nor a mature capsid, indicating that an intact viral membrane can substitute for the function of the viral capsid during reverse transcription in vitro. Collectively, these results demonstrate that the viral capsid functions as a nanoscale container for reverse transcription during HIV-1 infection.


Subject(s)
Capsid , HIV-1 , Reverse Transcription , HIV-1/physiology , HIV-1/metabolism , Capsid/metabolism , Humans , Virion/metabolism , HIV Envelope Protein gp41/metabolism , HIV Envelope Protein gp41/genetics , Virus Assembly/physiology , HIV Infections/virology , HIV Infections/metabolism , Phytic Acid/metabolism
6.
Nat Commun ; 15(1): 7702, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39231967

ABSTRACT

The human gut virome, which is mainly composed of bacteriophages, also includes viruses infecting archaea, yet their role remains poorly understood due to lack of isolates. Here, we characterize a temperate archaeal virus (MSTV1) infecting Methanobrevibacter smithii, the dominant methanogenic archaeon of the human gut. The MSTV1 genome is integrated in the host chromosome as a provirus which is sporadically induced, resulting in virion release. Using cryo-electron tomography, we capture several intracellular virion assembly intermediates and confirm that only a small fraction of the host population actively produces virions in vitro. Similar low frequency of induction is observed in a mouse colonization model, using mice harboring a stable consortium of 12 bacterial species (OMM12). Transcriptomic analysis suggests a regulatory lysogeny-lysis switch involving an interplay between viral proteins to maintain virus-host equilibrium, ensuring host survival and viral persistence. Thus, our study sheds light on archaeal virus-host interactions and highlights similarities with bacteriophages in establishing stable coexistence with their hosts in the gut.


Subject(s)
Archaeal Viruses , Gastrointestinal Microbiome , Methanobrevibacter , Animals , Humans , Methanobrevibacter/genetics , Methanobrevibacter/metabolism , Mice , Archaeal Viruses/genetics , Archaeal Viruses/physiology , Archaeal Viruses/ultrastructure , Genome, Viral/genetics , Virion/ultrastructure , Lysogeny , Female
7.
Science ; 385(6714): 1217-1224, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39264996

ABSTRACT

Chronic hepatitis B virus (HBV) infection poses a major global health challenge with massive morbidity and mortality. Despite a preventive vaccine, current treatments provide limited virus clearance, necessitating lifelong commitment. The HBV surface antigen (HBsAg) is crucial for diagnosis and prognosis, yet its high-resolution structure and assembly on the virus envelope remain elusive. Utilizing extensive datasets and advanced cryo-electron microscopy analysis, we present structural insights into HBsAg at a near-atomic resolution of 3.7 angstroms. HBsAg homodimers assemble into subviral particles with D2- and D4-like quasisymmetry, elucidating the dense-packing rules and structural adaptability of HBsAg. These findings provide insights into how HBsAg assembles into higher-order filaments and interacts with the capsid to form virions.


Subject(s)
Capsid , Hepatitis B Surface Antigens , Hepatitis B virus , Virion , Humans , Capsid/chemistry , Capsid/ultrastructure , Cryoelectron Microscopy , Hepatitis B Surface Antigens/chemistry , Hepatitis B virus/ultrastructure , Hepatitis B virus/chemistry , Hepatitis B virus/physiology , Protein Multimerization , Viral Envelope/chemistry , Viral Envelope/ultrastructure , Virion/ultrastructure , Virion/chemistry , Virus Assembly , Hepatitis B, Chronic/virology , Datasets as Topic
8.
Nat Commun ; 15(1): 8017, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39271696

ABSTRACT

The HIV-1 capsid is composed of capsid (CA) protein hexamers and pentamers (capsomers) that contain a central pore hypothesised to regulate capsid assembly and facilitate nucleotide import early during post-infection. These pore functions are mediated by two positively charged rings created by CA Arg-18 (R18) and Lys-25 (K25). Here we describe the forced evolution of viruses containing mutations in R18 and K25. Whilst R18 mutants fail to replicate, K25A viruses acquire compensating mutations that restore nearly wild-type replication fitness. These compensating mutations, which rescue reverse transcription and infection without reintroducing lost pore charges, map to three adaptation hot-spots located within and between capsomers. The second-site suppressor mutations act by restoring the formation of pentamers lost upon K25 mutation, enabling closed conical capsid assembly both in vitro and inside virions. These results indicate that there is no intrinsic requirement for K25 in either nucleotide import or capsid assembly. We propose that whilst HIV-1 must maintain a precise hexamer:pentamer equilibrium for proper capsid assembly, compensatory mutations can tune this equilibrium to restore fitness lost by mutation of the central pore.


Subject(s)
Capsid Proteins , Capsid , HIV-1 , Mutation , Virus Assembly , Virus Replication , HIV-1/genetics , HIV-1/physiology , Virus Assembly/genetics , Capsid Proteins/genetics , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Capsid/metabolism , Humans , Virus Replication/genetics , Virion/metabolism , Virion/genetics , HEK293 Cells , HIV Infections/virology , HIV Infections/genetics
9.
Viruses ; 16(9)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39339837

ABSTRACT

Hepatitis A virus (HAV) is an enteric virus mainly transmitted by the faecal-oral route. Belonging to the Picornaviridae family, HAV was first described as small naked particles, like all viruses of this family. However, for about a decade, it was demonstrated that HAV particles can exist surrounded by a lipid bilayer. This type of particle, called enveloped HAV (eHAV), acquires its lipid bilayer by hijacking a part of cell membranes during the virion egress in the last steps of the viral cycle. In vitro culture systems produce mainly eHAV, and so, to date, most of the studies on HAV have been carried out using this type of viral particle. In this study, a method based on lipid bilayer removal by chemical delipidation is proposed for the production of naked HAV particles. The resulting naked HAV particles conserve their infectivity and are therefore fully cultivable in vitro. By using this method, naked HAV particles can easily be produced in vitro and can be useful to perform further studies such as inactivation processes for the food industry, as HAV is a main concern for food safety.


Subject(s)
Hepatitis A virus , Virion , Hepatitis A virus/physiology , Humans , Virus Cultivation/methods , Animals , Cell Line , Hepatitis A/virology
10.
Viruses ; 16(9)2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39339894

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is a cancer-causing virus that establishes life-long infection. KSHV is implicated in the etiology of Kaposi's sarcoma, and a number of rare hematopoietic malignancies. The present study focuses on the KSHV open reading frame 20 (ORF20), a member of the conserved herpesvirus UL24 protein family containing five conserved homology domains and a conserved PD-(D/E)XK putative endonuclease motif, whose nuclease function has not been established to date. ORF20 encodes three co-linear protein isoforms, full length, intermediate, and short, though their differential functions are unknown. In an effort to determine the role of ORF20 during KSHV infection, we generated a recombinant ORF20-Null KSHV genome, which fails to express all three ORF20 isoforms. This genome was reconstituted in iSLK cells to establish a latent infection, which resulted in an accelerated transcription of viral mRNAs, an earlier accumulation of viral lytic proteins, an increase in the quantity of viral DNA copies, and a significant decrease in viral yield upon lytic reactivation. This was accompanied by early cell death of cells infected with the ORF20-Null virus. Functional complementation of the ORF20-Null mutant with the short ORF20 isoform rescued KSHV production, whereas its endonuclease mutant form failed to enhance lytic reactivation. Complementation with the short isoform further revealed a decrease in cell death as compared with ORF20-Null virus. Finally, expression of IL6 and CXCL8, previously shown to be affected by the hCMV UL24 homolog, was relatively low upon reactivation of cells infected with the ORF20-Null virus. These findings suggest that ORF20 protein, with its putative endonuclease motif, promotes coordinated lytic reactivation for increased infectious particle production.


Subject(s)
Herpesvirus 8, Human , Open Reading Frames , Viral Proteins , Virus Activation , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/physiology , Humans , Viral Proteins/genetics , Viral Proteins/metabolism , Cell Line , Virion/metabolism , Virion/genetics , Virus Replication , Virus Latency/genetics , DNA, Viral/genetics , Gene Expression Regulation, Viral , Genome, Viral
11.
Viruses ; 16(9)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39339899

ABSTRACT

HIV-1 virion maturation is an essential step in the viral replication cycle to produce infectious virus particles. Gag and Gag-Pol polyproteins are assembled at the plasma membrane of the virus-producer cells and bud from it to the extracellular compartment. The newly released progeny virions are initially immature and noninfectious. However, once the Gag polyprotein is cleaved by the viral protease in progeny virions, the mature capsid proteins assemble to form the fullerene core. This core, harboring two copies of viral genomic RNA, transforms the virion morphology into infectious virus particles. This morphological transformation is referred to as maturation. Virion maturation influences the distribution of the Env glycoprotein on the virion surface and induces conformational changes necessary for the subsequent interaction with the CD4 receptor. Several host factors, including proteins like cyclophilin A, metabolites such as IP6, and lipid rafts containing sphingomyelins, have been demonstrated to have an influence on virion maturation. This review article delves into the processes of virus maturation and Env glycoprotein recruitment, with an emphasis on the role of host cell factors and environmental conditions. Additionally, we discuss microscopic technologies for assessing virion maturation and the development of current antivirals specifically targeting this critical step in viral replication, offering long-acting therapeutic options.


Subject(s)
HIV-1 , Virion , Virus Assembly , Virus Replication , HIV-1/physiology , HIV-1/drug effects , Humans , Virion/metabolism , Virion/physiology , HIV Infections/virology , HIV Infections/drug therapy , Anti-HIV Agents/pharmacology , gag Gene Products, Human Immunodeficiency Virus/metabolism
12.
Viruses ; 16(8)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39205315

ABSTRACT

The efficiency of virus internalization into target cells is a major determinant of infectivity. SARS-CoV-2 internalization occurs via S-protein-mediated cell binding followed either by direct fusion with the plasma membrane or endocytosis and subsequent fusion with the endosomal membrane. Despite the crucial role of virus internalization, the precise kinetics of the processes involved remains elusive. We developed a pipeline, which combines live-cell microscopy and advanced image analysis, for measuring the rates of multiple internalization-associated molecular events of single SARS-CoV-2-virus-like particles (VLPs), including endosome ingression and pH change. Our live-cell imaging experiments demonstrate that only a few minutes after binding to the plasma membrane, VLPs ingress into RAP5-negative endosomes via dynamin-dependent scission. Less than two minutes later, VLP speed increases in parallel with a pH drop below 5, yet these two events are not interrelated. By co-imaging fluorescently labeled nucleocapsid proteins, we show that nucleocapsid release occurs with similar kinetics to VLP acidification. Neither Omicron mutations nor abrogation of the S protein polybasic cleavage site affected the rate of VLP internalization, indicating that they do not confer any significant advantages or disadvantages during this process. Finally, we observe that VLP internalization occurs two to three times faster in VeroE6 than in A549 cells, which may contribute to the greater susceptibility of the former cell line to SARS-CoV-2 infection. Taken together, our precise measurements of the kinetics of VLP internalization-associated processes shed light on their contribution to the effectiveness of SARS-CoV-2 propagation in cells.


Subject(s)
COVID-19 , Endosomes , SARS-CoV-2 , Virus Internalization , SARS-CoV-2/physiology , SARS-CoV-2/metabolism , Humans , Kinetics , COVID-19/virology , COVID-19/metabolism , Endosomes/metabolism , Endosomes/virology , Endocytosis , Animals , Hydrogen-Ion Concentration , Chlorocebus aethiops , Spike Glycoprotein, Coronavirus/metabolism , Vero Cells , Cell Membrane/metabolism , Cell Membrane/virology , Virion/metabolism
13.
Sci Signal ; 17(851): eadn8727, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39190708

ABSTRACT

Establishing a nonproductive, quiescent infection within monocytes is essential for the spread of human cytomegalovirus (HCMV). We investigated the mechanisms through which HCMV establishes a quiescent infection in monocytes. US28 is a virally encoded G protein-coupled receptor (GPCR) that is essential for silent infections within cells of the myeloid lineage. We found that preformed US28 was rapidly delivered to monocytes by HCMV viral particles, whereas the de novo synthesis of US28 was delayed for several days. A recombinant mutant virus lacking US28 (US28Δ) was unable to establish a quiescent infection, resulting in a fully productive lytic infection able to produce progeny virus. Infection with US28Δ HCMV resulted in the phosphorylation of the serine and threonine kinase Akt at Ser473 and Thr308, in contrast with the phosphorylation of Akt only at Ser473 after WT viral infection. Inhibiting the dual phosphorylation of Akt prevented the lytic replication of US28Δ, and ectopic expression of a constitutively phosphorylated Akt variant triggered lytic replication of wild-type HCMV. Mechanistically, we found that US28 was necessary and sufficient to attenuate epidermal growth factor receptor (EGFR) signaling induced during the entry of WT virus, which led to the site-specific phosphorylation of Akt at Ser473. Thus, particle-delivered US28 fine-tunes Akt activity by limiting HCMV-induced EGFR activation during viral entry, enabling quiescent infection in monocytes.


Subject(s)
Cytomegalovirus , ErbB Receptors , Monocytes , Proto-Oncogene Proteins c-akt , Viral Proteins , Virus Replication , Cytomegalovirus/physiology , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Humans , Monocytes/virology , Monocytes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Phosphorylation , Viral Proteins/metabolism , Viral Proteins/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , Virion/metabolism , Virion/genetics , Receptors, Chemokine/metabolism , Receptors, Chemokine/genetics , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/genetics , Signal Transduction
14.
Hum Gene Ther ; 35(17-18): 604-616, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39150015

ABSTRACT

The invention of next-generation CRISPR/Cas gene editing tools, like base and prime editing, for correction of gene variants causing disease, has created hope for in vivo use in patients leading to wider clinical translation. To realize this potential, delivery vehicles that can ferry gene editing tool kits safely and effectively into specific cell populations or tissues are in great demand. In this review, we describe the development of enveloped retrovirus-derived particles as carriers of "ready-to-work" ribonucleoprotein complexes consisting of Cas9-derived editor proteins and single guide RNAs. We present arguments for adapting viruses for cell-targeted protein delivery and describe the status after a decade-long development period, which has already shown effective editing in primary cells, including T cells and hematopoietic stem cells, and in tissues targeted in vivo, including mouse retina, liver, and brain. Emerging evidence has demonstrated that engineered virus-derived nanoparticles can accommodate both base and prime editors and seems to fertilize a sprouting hope that such particles can be further developed and produced in large scale for therapeutic applications.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Ribonucleoproteins , Gene Editing/methods , Humans , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Animals , Virion/metabolism , Virion/genetics , Retroviridae/genetics , Genetic Vectors/genetics , Genetic Therapy/methods , Gene Transfer Techniques , RNA, Guide, CRISPR-Cas Systems/genetics
15.
Int J Biol Macromol ; 278(Pt 4): 134839, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39163959

ABSTRACT

The virus is the smallest known replicative unit, usually in nanometer-range sizes. The most simple and sensitive detection assay involves molecular amplification of nucleic acids. This work shows a novel, straightforward detection based on the interaction of viral particles with fluorescent nanoconstructs without using enzymatic amplification, washing or separation steps. Fluorescent nanoconstructs are prepared with individual quantum dots of different emitting green and red fluorescence as a core. They are decorated with aptamers developed to recognise the receptor-binding region of the SARS-CoV-2 spike protein. Nanoconstructs can recognise SARS-CoV-2 viral particles fixed onto a coverglass generating aggregates. Meanwhile, SARS-CoV-2 viral particles/nanoconstructs complexes in solution yield aggregates and complexes, which a fluorescence microscope can visualise. The multiple molecular recognition allowed the detection of SARS-CoV-2 viral particles from a few microliters of patient swabs. This specific SARS-CoV-2/nanoconstructs interaction generates insoluble and precipitating aggregates. By using a mixture of green and red fluorescent nanoconstructs, upon the viral particle interaction, they yield heterochromatic green, red and yellow spectral fluorescence, easily identifiable by a fluorescence microscope. Washing and separation steps are not required, and aggregates allow one to easily recognise them, offering a sensitive, simple, and cheap alternative for viral detection.


Subject(s)
Aptamers, Nucleotide , COVID-19 , Microscopy, Fluorescence , Quantum Dots , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virion , SARS-CoV-2/isolation & purification , Quantum Dots/chemistry , Humans , Aptamers, Nucleotide/chemistry , Virion/isolation & purification , COVID-19/virology , COVID-19/diagnosis , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry
16.
J Virol ; 98(9): e0068024, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39158347

ABSTRACT

Betacoronaviruses encode a conserved accessory gene within the +1 open reading frame (ORF) of nucleocapsid called the internal N gene. This gene is referred to as "I" for mouse hepatitis virus (MHV), ORF9b for severe acute respiratory CoV (SARS-CoV) and SARS-CoV-2, and ORF8b for Middle East respiratory syndrome CoV (MERS-CoV). Previous studies have shown ORF8b and ORF9b have immunoevasive properties, while the only known information for MHV I is its localization within the virion of the hepatotropic/neurotropic A59 strain of MHV. Whether MHV I is an innate immune antagonist or has other functions has not been evaluated. In this report, we show that the I protein of the neurotropic JHM strain of MHV (JHMV) lacks a N terminal domain present in other MHV strains, has immunoevasive properties, and is a component of the virion. Genetic deletion of JHMV I (rJHMVIΔ57-137) resulted in a highly attenuated virus both in vitro and in vivo that displayed a post RNA replication/transcription defect that ultimately resulted in fewer infectious virions packaged compared with wild-type virus. This phenotype was only seen for rJHMVIΔ57-137, suggesting the structural changes predicted for A59 I altered its function, as genetic deletion of A59 I did not change viral replication or pathogenicity. Together, these data show that JHMV I both acts as a mild innate immune antagonist and aids in viral assembly and infectious virus production, and suggest that the internal N proteins from different betacoronaviruses have both common and virus strain-specific properties.IMPORTANCECoV accessory genes are largely studied in overexpression assays and have been identified as innate immune antagonists. However, functions identified after overexpression are often not confirmed in the infected animal host. Furthermore, some accessory proteins are components of the CoV virion, but their role in viral replication and release remains unclear. Here, we utilized reverse genetics to abrogate expression of a conserved CoV accessory gene, the internal N ("I") gene, of the neurotropic JHMV strain of MHV and found that loss of the I gene resulted in a post replication defect that reduced virion assembly and ultimately infectious virus production, while also increasing some inflammatory molecule expression. Thus, the JHMV I protein has roles in virion assembly that were previously underappreciated and in immunoevasion.


Subject(s)
Murine hepatitis virus , Viral Proteins , Virus Replication , Murine hepatitis virus/genetics , Murine hepatitis virus/pathogenicity , Murine hepatitis virus/immunology , Murine hepatitis virus/physiology , Animals , Mice , Virulence , Viral Proteins/metabolism , Viral Proteins/genetics , Virion/metabolism , Immunity, Innate , Coronavirus Infections/virology , Coronavirus Infections/immunology , Cell Line , Open Reading Frames , Humans
17.
ACS Synth Biol ; 13(9): 2710-2717, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39178386

ABSTRACT

Recombinant adeno-associated viruses (rAAVs) have emerged as important tools for gene therapy and, more recently, vaccine development. Nonetheless, manufacturing can be costly and time-consuming, emphasizing the importance of alternative production platforms. We investigate the potential of E. coli-based cell-free protein synthesis (CFPS) to produce recombinant AAV5 virus-like particles (VLPs). AAV5 virus protein 3 (VP3) constructs, both with and without Strep-tag II, were expressed with CFPS. Lower reaction temperatures resulted in increased solubility, with the untagged variant containing nearly 90% more soluble VLP VP3 protein at 18 °C than at 37 °C. Affinity chromatography of N-terminally Strep(II)-tagged VP3 enabled successful isolation with minimal processing. DLS and TEM confirmed the presence of ∼20 nm particles. Furthermore, the N-terminally tagged AAV5 VP3 VLPs were biologically active, successfully internalizing into HeLa cells. This study describes an innovative approach to AAV VLP production using E. coli-based CFPS, demonstrating its potential for rapid and biologically active AAV VLP synthesis.


Subject(s)
Cell-Free System , Dependovirus , Escherichia coli , Escherichia coli/genetics , Escherichia coli/metabolism , Dependovirus/genetics , Humans , HeLa Cells , Capsid Proteins/genetics , Capsid Proteins/metabolism , Capsid Proteins/biosynthesis , Protein Biosynthesis , Virion/genetics , Virion/metabolism
18.
Arch Virol ; 169(8): 172, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39096433

ABSTRACT

Goatpox and sheeppox are highly contagious and economically important viral diseases of small ruminants. Due to the risk they pose to animal health, livestock production, and international trade, capripoxviruses are a considerable threat to the livestock economy. In this study, we expressed two core proteins (A4L and A12L) and one extracellular enveloped virion protein (A33R) of goatpox virus in a baculovirus expression vector system and evaluated their use as diagnostic antigens in ELISA. Full-length A4L, A12L, and A33R genes of the GTPV Uttarkashi strain were amplified, cloned into the pFastBac HT A donor vector, and introduced into DH10Bac cells containing a baculovirus shuttle vector plasmid to generate recombinant bacmids. The recombinant baculoviruses were produced in Sf-21 cells by transfection, and proteins were expressed in TN5 insect cells. The recombinant proteins were analysed by SDS-PAGE and confirmed by western blot, with expected sizes of ~30 kDa, ~31 kDa, and ~32 kDa for A4L, A12L, and A33R, respectively. The recombinant proteins were purified, and the immunoreactivity of the purified proteins was confirmed by western blot using anti-GTPV serum. The antigenic specificity of the expressed proteins as diagnostic antigens was evaluated by testing their reactivity with infected, vaccinated, and negative GTPV/SPPV serum in indirect ELISA, and the A33R-based indirect ELISA was optimized. The diagnostic sensitivity and specificity of the A33R-based indirect ELISA were found to be of 89% and 94% for goats and 98% and 91%, for sheep, respectively. No cross-reactivity was observed with other related viruses. The recombinant-A33R-based indirect ELISA developed in the present study shows that it has potential for the detection of antibodies in GTPV and SPPV infected/vaccinated animals.


Subject(s)
Baculoviridae , Capripoxvirus , Enzyme-Linked Immunosorbent Assay , Goat Diseases , Goats , Viral Envelope Proteins , Capripoxvirus/genetics , Capripoxvirus/isolation & purification , Baculoviridae/genetics , Animals , Goat Diseases/virology , Goat Diseases/diagnosis , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Goats/virology , Enzyme-Linked Immunosorbent Assay/methods , Poxviridae Infections/diagnosis , Poxviridae Infections/veterinary , Poxviridae Infections/virology , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/immunology , Virion/genetics , Viral Core Proteins/genetics , Viral Core Proteins/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Sf9 Cells , Antigens, Viral/genetics , Antigens, Viral/immunology , Cell Line , Gene Expression
19.
ACS Nano ; 18(32): 21024-21037, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39087909

ABSTRACT

Virus-like particles (VLPs) have untapped potential for packaging and delivery of macromolecular cargo. To be a broadly useful platform, there needs to be a strategy for attaching macromolecules to the inside or the outside of the VLP with minimal modification of the platform or cargo. Here, we repurpose antiviral compounds that bind to hepatitis B virus (HBV) capsids to create a chemical tag to noncovalently attach cargo to the VLP. Our tag consists of a capsid assembly modulator, HAP13, connected to a linker terminating in maleimide. Our cargo is a green fluorescent protein (GFP) with a single addressable cysteine, a feature that can be engineered in many proteins. The HAP-GFP construct maintained HAP's intrinsic ability to bind HBV capsids and accelerate assembly. We investigated the capacity of HAP-GFP to coassemble with HBV capsid protein and bind to preassembled capsids. HAP-GFP binding was concentration-dependent, sensitive to capsid stability, and dependent on linker length. Long linkers had the greatest activity to bind capsids, while short linkers impeded assembly and damaged intact capsids. In coassembly reactions, >20 HAP-GFP molecules were presented on the outside and inside of the capsid, concentrating the cargo by more than 100-fold compared to bulk solution. We also tested an HAP-GFP with a cleavable linker so that external GFP molecules could be removed, resulting in exclusive internal packaging. These results demonstrate a generalizable strategy for attaching cargo to a VLP, supporting development of HBV as a modular VLP platform.


Subject(s)
Capsid , Green Fluorescent Proteins , Hepatitis B virus , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/chemistry , Capsid/chemistry , Capsid/metabolism , Virus Assembly , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Virion/metabolism , Virion/chemistry , Surface Properties
20.
Methods Mol Biol ; 2838: 77-89, 2024.
Article in English | MEDLINE | ID: mdl-39126624

ABSTRACT

Epizootic hemorrhagic disease virus (EHDV), like other orbiviruses, infects and replicates in mammalian and insect vector cells. Within its ruminant hosts EHDV, like bluetongue virus (BTV), it has mainly been associated with infection of endothelial cells of capillaries as well as leukocyte subsets. Furthermore, EHDV infects and replicates within its biological vector, Culicoides biting midges and Culicoides-derived cells. A wide range of common laboratory cell lines such as BHK, BSR, and Vero cells are susceptible to infection with certain EHDV strains. Cell culture supernatants of infected cells are commonly used for both in vivo and in vitro infection studies. For specific virological or immunological studies, using highly purified virus particles, however, might be beneficial or even required. Here we describe a purification method for EHDV particles, which had been originally developed for certain strains of BTV.


Subject(s)
Hemorrhagic Disease Virus, Epizootic , Virion , Animals , Hemorrhagic Disease Virus, Epizootic/isolation & purification , Cell Line , Virion/isolation & purification , Chlorocebus aethiops , Vero Cells , Orbivirus/isolation & purification , Ceratopogonidae/virology , Insecta/virology , Reoviridae Infections/virology , Reoviridae Infections/veterinary , Cricetinae
SELECTION OF CITATIONS
SEARCH DETAIL