Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Mol Biol ; 435(16): 168182, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37328094

ABSTRACT

Biomolecular condensates (BMCs) play important roles incellular structures includingtranscription factories, splicing speckles, and nucleoli. BMCs bring together proteins and other macromolecules, selectively concentrating them so that specific reactions can occur without interference from the surrounding environment. BMCs are often made up of proteins that contain intrinsically disordered regions (IDRs), form phase-separated spherical puncta, form liquid-like droplets that undergo fusion and fission, contain molecules that are mobile, and are disrupted with phase-dissolving drugs such as 1,6-hexanediol. In addition to cellular proteins, many viruses, including influenza A, SARS-CoV-2, and human immunodeficiency virus type 1 (HIV-1) encode proteins that undergo phase separation and rely on BMC formation for replication. In prior studies of the retrovirus Rous sarcoma virus (RSV), we observed that the Gag protein forms discrete spherical puncta in the nucleus, cytoplasm, and at the plasma membrane that co-localize with viral RNA and host factors, raising the possibility that RSV Gag forms BMCs that participate in the intracellular phase of the virion assembly pathway. In our current studies, we found that Gag contains IDRs in the N-terminal (MAp2p10) and C-terminal (NC) regions of the protein and fulfills many criteria of BMCs. Although the role of BMC formation in RSV assembly requires further study, our results suggest the biophysical properties of condensates are required for the formation of Gag complexes in the nucleus and the cohesion of these complexes as they traffic through the nuclear pore, into the cytoplasm, and to the plasma membrane, where the final assembly and release of virus particles occurs.


Subject(s)
Biomolecular Condensates , Gene Products, gag , Intrinsically Disordered Proteins , Rous sarcoma virus , Humans , Biomolecular Condensates/metabolism , Biomolecular Condensates/virology , Gene Products, gag/chemistry , Gene Products, gag/metabolism , Rous sarcoma virus/metabolism , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Phase Transition
2.
J Virol ; 95(20): e0064821, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34319154

ABSTRACT

During retroviral replication, unspliced viral genomic RNA (gRNA) must escape the nucleus for translation into viral proteins and packaging into virions. "Complex" retroviruses, such as human immunodeficiency virus (HIV), use cis-acting elements on the unspliced gRNA in conjunction with trans-acting viral proteins to facilitate this escape. "Simple" retroviruses, such as Mason-Pfizer monkey virus (MPMV) and murine leukemia virus (MLV), exclusively use cis-acting elements on the gRNA in conjunction with host nuclear export proteins for nuclear escape. Uniquely, the simple retrovirus Rous sarcoma virus (RSV) has a Gag structural protein that cycles through the nucleus prior to plasma membrane binding. This trafficking has been implicated in facilitating gRNA nuclear export and is thought to be a required mechanism. Previously described mutants that abolish nuclear cycling displayed enhanced plasma membrane binding, enhanced virion release, and a significant loss in genome incorporation resulting in loss of infectivity. Here, we describe a nuclear cycling-deficient RSV Gag mutant that has similar plasma membrane binding and genome incorporation to wild-type (WT) virus and surprisingly is replication competent, albeit with a slower rate of spread than observed in WT virus. This mutant suggests that RSV Gag nuclear cycling is not strictly required for RSV replication. IMPORTANCE While mechanisms for retroviral Gag assembly at the plasma membrane are beginning to be characterized, characterization of intermediate trafficking locales remain elusive. This is in part due to the difficulty of tracking individual proteins from translation to plasma membrane binding. Rous sarcoma virus (RSV) Gag nuclear cycling is a unique phenotype that may provide comparative insight to viral trafficking evolution and may present a model intermediate to cis- and trans-acting mechanisms for gRNA export.


Subject(s)
Active Transport, Cell Nucleus/physiology , Gene Products, gag/genetics , Rous sarcoma virus/genetics , Active Transport, Cell Nucleus/genetics , Animals , Cell Line , Cell Nucleus/virology , Gene Products, gag/metabolism , Genome, Viral/genetics , Humans , Mice , RNA, Viral/genetics , Retroviridae/genetics , Rous sarcoma virus/metabolism , Virion/metabolism , Virus Assembly
3.
mBio ; 11(2)2020 04 07.
Article in English | MEDLINE | ID: mdl-32265329

ABSTRACT

Packaging of genomic RNA (gRNA) by retroviruses is essential for infectivity, yet the subcellular site of the initial interaction between the Gag polyprotein and gRNA remains poorly defined. Because retroviral particles are released from the plasma membrane, it was previously thought that Gag proteins initially bound to gRNA in the cytoplasm or at the plasma membrane. However, the Gag protein of the avian retrovirus Rous sarcoma virus (RSV) undergoes active nuclear trafficking, which is required for efficient gRNA encapsidation (L. Z. Scheifele, R. A. Garbitt, J. D. Rhoads, and L. J. Parent, Proc Natl Acad Sci U S A 99:3944-3949, 2002, https://doi.org/10.1073/pnas.062652199; R. Garbitt-Hirst, S. P. Kenney, and L. J. Parent, J Virol 83:6790-6797, 2009, https://doi.org/10.1128/JVI.00101-09). These results raise the intriguing possibility that the primary contact between Gag and gRNA might occur in the nucleus. To examine this possibility, we created a RSV proviral construct that includes 24 tandem repeats of MS2 RNA stem-loops, making it possible to track RSV viral RNA (vRNA) in live cells in which a fluorophore-conjugated MS2 coat protein is coexpressed. Using confocal microscopy, we observed that both wild-type Gag and a nuclear export mutant (Gag.L219A) colocalized with vRNA in the nucleus. In live-cell time-lapse images, the wild-type Gag protein trafficked together with vRNA as a single ribonucleoprotein (RNP) complex in the nucleoplasm near the nuclear periphery, appearing to traverse the nuclear envelope into the cytoplasm. Furthermore, biophysical imaging methods suggest that Gag and the unspliced vRNA physically interact in the nucleus. Taken together, these data suggest that RSV Gag binds unspliced vRNA to export it from the nucleus, possibly for packaging into virions as the viral genome.IMPORTANCE Retroviruses cause severe diseases in animals and humans, including cancer and acquired immunodeficiency syndromes. To propagate infection, retroviruses assemble new virus particles that contain viral proteins and unspliced vRNA to use as gRNA. Despite the critical requirement for gRNA packaging, the molecular mechanisms governing the identification and selection of gRNA by the Gag protein remain poorly understood. In this report, we demonstrate that the Rous sarcoma virus (RSV) Gag protein colocalizes with unspliced vRNA in the nucleus in the interchromatin space. Using live-cell confocal imaging, RSV Gag and unspliced vRNA were observed to move together from inside the nucleus across the nuclear envelope, suggesting that the Gag-gRNA complex initially forms in the nucleus and undergoes nuclear export into the cytoplasm as a viral ribonucleoprotein (vRNP) complex.


Subject(s)
Cell Nucleus/virology , Gene Products, gag/metabolism , Genome, Viral , RNA, Viral/metabolism , Rous sarcoma virus/genetics , Virus Assembly , Active Transport, Cell Nucleus , Animals , Cell Line , Cell Line, Transformed , Cell Nucleus/metabolism , Fibroblasts/virology , Microscopy, Confocal , Quail , RNA, Viral/analysis , Rous sarcoma virus/metabolism , Time-Lapse Imaging
4.
Viruses ; 9(8)2017 08 01.
Article in English | MEDLINE | ID: mdl-28763028

ABSTRACT

All retroviruses use their full-length primary transcript as the major mRNA for Group-specific antigen (Gag) capsid proteins. This results in a long 3' untranslated region (UTR) downstream of the termination codon. In the case of Rous sarcoma virus (RSV), there is a 7 kb 3'UTR downstream of the gag terminator, containing the pol, env, and src genes. mRNAs containing long 3'UTRs, like those with premature termination codons, are frequently recognized by the cellular nonsense-mediated mRNA decay (NMD) machinery and targeted for degradation. To prevent this, RSV has evolved an RNA stability element (RSE) in the RNA immediately downstream of the gag termination codon. This 400-nt RNA sequence stabilizes premature termination codons (PTCs) in gag. It also stabilizes globin mRNAs with long 3'UTRs, when placed downstream of the termination codon. It is not clear how the RSE stabilizes the mRNA and prevents decay. We show here that the presence of RSE inhibits deadenylation severely. In addition, the RSE also impairs decapping (DCP2) and 5'-3' exonucleolytic (XRN1) function in knockdown experiments in human cells.


Subject(s)
3' Untranslated Regions , RNA Stability , RNA, Messenger/genetics , RNA, Viral/genetics , Rous sarcoma virus/genetics , Codon, Terminator , Gene Knockdown Techniques , Gene Products, gag/genetics , Gene Products, gag/metabolism , Humans , Nonsense Mediated mRNA Decay , Protein Biosynthesis , RNA, Messenger/metabolism , RNA, Viral/metabolism , Rous sarcoma virus/metabolism
5.
Proc Natl Acad Sci U S A ; 114(26): E5148-E5157, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28607078

ABSTRACT

The extent of virus transmission among individuals and species is generally determined by the presence of specific membrane-embedded virus receptors required for virus entry. Interaction of the viral envelope glycoprotein (Env) with a specific cellular receptor is the first and crucial step in determining host specificity. Using a well-established retroviral model-avian Rous sarcoma virus (RSV)-we analyzed changes in an RSV variant that had repeatedly been able to infect rodents. By envelope gene (env) sequencing, we identified eight mutations that do not match the already described mutations influencing the host range. Two of these mutations-one at the beginning (D32G) of the surface Env subunit (SU) and the other at the end of the fusion peptide region (L378S)-were found to be of critical importance, ensuring transmission to rodent, human, and chicken cells lacking the appropriate receptor. Furthermore, we carried out assays to examine the virus entry mechanism and concluded that these two mutations cause conformational changes in the Env variant and that these changes lead to an activated, or primed, state of Env (normally induced after Env interaction with the receptor). In summary, our results indicate that retroviral host range extension is caused by spontaneous Env activation, which circumvents the need for original cell receptor. This activation is, in turn, caused by mutations in various env regions.


Subject(s)
Gene Products, env , Genetic Vectors , Mutation, Missense , Rous sarcoma virus , Transduction, Genetic , Amino Acid Substitution , Animals , Cell Line, Tumor , Chickens , Gene Products, env/genetics , Gene Products, env/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Rats , Rous sarcoma virus/genetics , Rous sarcoma virus/metabolism
6.
Mol Ther ; 25(5): 1187-1198, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28365028

ABSTRACT

Conventional plasmid vectors are incapable of achieving sustained levels of transgene expression in vivo even in quiescent mammalian tissues because the transgene expression cassette is silenced. Transcriptional silencing results from the presence of the bacterial plasmid backbone or virtually any DNA sequence of >1 kb in length placed outside of the expression cassette. Here, we show that transcriptional silencing can be substantially forestalled by increasing the An/Tn sequence composition in the plasmid bacterial backbone. Increasing numbers of An/Tn sequences increased sustained transcription of both backbone sequences and adjacent expression cassettes. In order to recapitulate these expression profiles in compact and portable plasmid DNA backbones, we engineered the standard kanamycin or ampicillin antibiotic resistance genes, optimizing the number of An/Tn sequence without altering the encoded amino acids. The resulting vector backbones yield sustained transgene expression from mouse liver, providing generic DNA vectors capable of sustained transgene expression without additional genes or mammalian regulatory elements.


Subject(s)
Dependovirus/genetics , Drug Resistance, Microbial/genetics , Genetic Engineering/methods , Genetic Vectors/chemistry , Plasmids/chemistry , alpha 1-Antitrypsin/genetics , Ampicillin/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Dependovirus/metabolism , Female , Gene Silencing , Genetic Vectors/metabolism , Humans , Kanamycin/pharmacology , Liver/metabolism , Liver/virology , Mice , Mice, Inbred C57BL , Nucleotide Motifs , Plasmids/metabolism , Promoter Regions, Genetic/drug effects , Rous sarcoma virus/genetics , Rous sarcoma virus/metabolism , Transcription, Genetic/drug effects , Transgenes , alpha 1-Antitrypsin/metabolism
7.
Virology ; 498: 181-191, 2016 11.
Article in English | MEDLINE | ID: mdl-27596537

ABSTRACT

The weak polyadenylation site (PAS) of Rous sarcoma virus (RSV) is activated by the juxtaposition of SR protein binding sites within the spatially separate negative regulator of splicing (NRS) element and the env RNA splicing enhancer (Env enhancer), which are far upstream of the PAS. Juxtaposition occurs by formation of the NRS - 3' ss splicing regulatory complex and is thought to provide a threshold of SR proteins that facilitate long-range stimulation of the PAS. We provide evidence for the threshold model by showing that greater than three synthetic SR protein binding sites are needed to substitute for the Env enhancer, that either the NRS or Env enhancer alone promotes polyadenylation when the distance to the PAS is decreased, and that SR protein binding sites promote binding of the polyadenylation factor cleavage factor I (CFIm) to the weak PAS. These observations may be relevant for cellular PASs.


Subject(s)
Polyadenylation , RNA, Messenger , RNA, Viral , RNA-Binding Proteins/metabolism , Rous sarcoma virus/genetics , Rous sarcoma virus/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Binding Sites , Cell Line , Gene Order , Open Reading Frames , Poly A , Protein Binding
8.
Nature ; 530(7590): 362-6, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26887497

ABSTRACT

Integration of the reverse-transcribed viral DNA into the host genome is an essential step in the life cycle of retroviruses. Retrovirus integrase catalyses insertions of both ends of the linear viral DNA into a host chromosome. Integrase from HIV-1 and closely related retroviruses share the three-domain organization, consisting of a catalytic core domain flanked by amino- and carboxy-terminal domains essential for the concerted integration reaction. Although structures of the tetrameric integrase-DNA complexes have been reported for integrase from prototype foamy virus featuring an additional DNA-binding domain and longer interdomain linkers, the architecture of a canonical three-domain integrase bound to DNA remained elusive. Here we report a crystal structure of the three-domain integrase from Rous sarcoma virus in complex with viral and target DNAs. The structure shows an octameric assembly of integrase, in which a pair of integrase dimers engage viral DNA ends for catalysis while another pair of non-catalytic integrase dimers bridge between the two viral DNA molecules and help capture target DNA. The individual domains of the eight integrase molecules play varying roles to hold the complex together, making an extensive network of protein-DNA and protein-protein contacts that show both conserved and distinct features compared with those observed for prototype foamy virus integrase. Our work highlights the diversity of retrovirus intasome assembly and provides insights into the mechanisms of integration by HIV-1 and related retroviruses.


Subject(s)
DNA, Viral/chemistry , Integrases/chemistry , Rous sarcoma virus/chemistry , Rous sarcoma virus/enzymology , Catalytic Domain , Crystallography, X-Ray , DNA, Viral/metabolism , HIV-1/enzymology , HIV-1/metabolism , Integrases/metabolism , Models, Molecular , Protein Binding , Protein Multimerization , Rous sarcoma virus/genetics , Rous sarcoma virus/metabolism , Spumavirus/enzymology , Virus Integration
9.
Pharm Res ; 32(11): 3699-707, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26047779

ABSTRACT

PURPOSE: Virus-like particles (VLPs) have been used as drug carriers for drug delivery systems. In this study, hCC49 single chain fragment variable (scFv)-displaying Rous sarcoma virus-like particles (RSV VLPs) were produced in silkworm larvae to be a specific carrier of an anti-cancer drug. METHOD: RSV VLPs displaying hCC49 scFv were created by the fusion of the transmembrane and cytoplasmic domains of hemagglutinin from influenza A (H1N1) virus and produced in silkworm larvae. The display of hCC49 scFv on the surface of RSV VLPs was confirmed by enzyme-linked immunosorbent assay using tumor-associated glycoprotein-72 (TAG-72), fluorescent microscopy, and immunoelectron microscopy. Fluorescein isothiocyanate (FITC) or doxorubicin (DOX) was incorporated into hCC49 scFv-displaying RSV VLPs by electroporation and specific targeting of these VLPs was investigated by fluorescent microscopy and cytotoxicity assay using LS174T cells. RESULTS: FITC was delivered to LS174T human colon adenocarcinoma cells by hCC49 scFv-displaying RSV VLPs, but not by RSV VLPs. This indicated that hCC49 scFv allowed FITC-loaded RSV VLPs to be delivered to LS174T cells. DOX, which is an anti-cancer drug with intrinsic red fluorescence, was also loaded into hCC49 scFv-displaying RSV VLPs by electroporation; the DOX-loaded hCC49 scFv-displaying RSV VLPs killed LS174T cells via the specific delivery of DOX that was mediated by hCC49 scFv. HEK293 cells were alive even though in the presence of DOX-loaded hCC49 scFv-displaying RSV VLPs. CONCLUSION: These results showed that hCC49 scFv-displaying RSV VLPs from silkworm larvae offered specific drug delivery to colon carcinoma cells in vitro. This scFv-displaying enveloped VLP system could be applied to drug and gene delivery to other target cells.


Subject(s)
Antibodies, Neoplasm/genetics , Antineoplastic Agents/administration & dosage , Drug Delivery Systems/methods , Rous sarcoma virus/genetics , Single-Chain Antibodies/genetics , Virion/genetics , Animals , Bombyx/genetics , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/pathology , Drug Carriers , Gene Products, gag/metabolism , HEK293 Cells , Humans , Larva/genetics , Rous sarcoma virus/metabolism , Vaccines, Virus-Like Particle/genetics , Virion/metabolism
10.
Mol Brain ; 8: 12, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25887710

ABSTRACT

BACKGROUND: Viral vectors are frequently used to deliver and direct expression of transgenes in a spatially and temporally restricted manner within the nervous system of numerous model organisms. Despite the common use of viral vectors to direct ectopic expression of transgenes within the nervous system, creating high titer viral vectors that are capable of expressing very large transgenes or difficult to express transgenes imposes unique challenges. Here we describe the development of adeno-associated viruses (AAV) and lentiviruses designed to express the large and difficult to express GluN2A or GluN2B subunits of the N-methyl-D-aspartate receptor (NMDA) receptor, specifically within neurons. RESULTS: We created a number of custom designed AAV and lentiviral vectors that were optimized for large transgenes, by minimizing DNA sequences that were not essential, utilizing short promoter sequences of 8 widely used promoters (RSV, EFS, TRE3G, 0.4αCaMKII, 1.3αCaMKII, 0.5Synapsin, 1.1Synapsin and CMV) and utilizing a very short (~75 bps) 3' untranslated sequence. Not surprisingly these promoters differed in their ability to express the GluN2 subunits, however surprisingly we found that the neuron specific synapsin and αCaMKII, promoters were incapable of conferring detectable expression of full length GluN2 subunits and detectable expression could only be achieved from these promoters if the transgene included an intron or if the GluN2 subunit transgenes were truncated to only include the coding regions of the GluN2 transmembrane domains. CONCLUSIONS: We determined that viral packaging limit, transgene promoter and the presence of an intron within the transgene were all important factors that contributed to being able to successfully develop viral vectors designed to deliver and express GluN2 transgenes in a neuron specific manner. Because these vectors have been optimized to accommodate large open reading frames and in some cases contain an intron to facilitate expression of difficult to express transgenes, these viral vectors likely could be useful for delivering and expressing many large or difficult to express transgenes in a neuron specific manner.


Subject(s)
Genetic Vectors/metabolism , Lentivirus/metabolism , Neurons/metabolism , Transgenes , Animals , Dependovirus/metabolism , Genome, Viral , Green Fluorescent Proteins/metabolism , Introns/genetics , Male , Mice, Inbred C57BL , Mutant Proteins/metabolism , Plasmids/metabolism , Promoter Regions, Genetic , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Rous sarcoma virus/metabolism
11.
J Virol ; 87(24): 13598-608, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24109216

ABSTRACT

In most retroviruses, plasma membrane (PM) association of the Gag structural protein is a critical step in viral assembly, relying in part on interaction between the highly basic Gag MA domain and the negatively charged inner leaflet of the PM. Assembly is thought to begin with Gag dimerization followed by multimerization, resulting in a hexameric lattice. To directly address the role of multimerization in membrane binding, we fused the MA domains of Rous sarcoma virus (RSV) and HIV-1 to the chemically inducible dimerization domain FK506-binding protein (FKBP) or to the hexameric protein CcmK4 from cyanobacteria. The cellular localization of the resulting green fluorescent protein (GFP)-tagged chimeric proteins was examined by fluorescence imaging, and the association of the proteins with liposomes was quantified by flotation in sucrose gradients, following synthesis in a reticulocyte extract or as purified proteins. Four lipid compositions were tested, representative of liposomes commonly reported in flotation experiments. By themselves, GFP-tagged RSV and HIV-1 MA proteins were largely cytoplasmic, but both hexamerized proteins were highly concentrated at the PM. Dimerization led to partial PM localization for HIV-1 MA. These in vivo effects of multimerization were reproduced in vitro. In flotation analyses, the intact RSV and HIV-1 Gag proteins were more similar to multimerized MA than to monomeric MA. RNA is reported to compete with acidic liposomes for HIV-1 Gag binding, and thus we also examined the effects of RNase treatment or tRNA addition on flotation. tRNA competed with liposomes in the case of some but not all lipid compositions and ionic strengths. Taken together, our results further underpin the model that multimerization is critical for PM association of retroviral Gag proteins. In addition, they suggest that the modulation of membrane binding by RNA, as previously reported for HIV-1, may not hold for RSV.


Subject(s)
Cell Membrane/virology , Gene Products, gag/chemistry , Gene Products, gag/metabolism , HIV Infections/virology , HIV-1/metabolism , Rous sarcoma virus/metabolism , Sarcoma, Avian/virology , gag Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Cell Line , Cytoplasm/virology , Gene Products, gag/genetics , HIV-1/chemistry , HIV-1/genetics , Humans , Protein Multimerization , Protein Structure, Tertiary , Quail , Rous sarcoma virus/chemistry , Rous sarcoma virus/genetics , gag Gene Products, Human Immunodeficiency Virus/genetics
12.
J Virol ; 87(24): 13655-64, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24109217

ABSTRACT

Retrovirus maturation involves sequential cleavages of the Gag polyprotein, initially arrayed in a spherical shell, leading to formation of capsids with polyhedral or conical morphology. Evidence suggests that capsids assemble de novo inside maturing virions from dissociated capsid (CA) protein, but the possibility persists of a displacive pathway in which the CA shell remains assembled but is remodeled. Inhibition of the final cleavage between CA and spacer peptide SP1/SP blocks the production of mature capsids. We investigated whether retention of SP might render CA assembly incompetent by testing the ability of Rous sarcoma virus (RSV) CA-SP to assemble in vitro into icosahedral capsids. Capsids were indeed assembled and were indistinguishable from those formed by CA alone, indicating that SP was disordered. We also used cryo-electron tomography to characterize HIV-1 particles produced in the presence of maturation inhibitor PF-46396 or with the cleavage-blocking CA5 mutation. Inhibitor-treated virions have a shell that resembles the CA layer of the immature Gag shell but is less complete. Some CA protein is generated but usually not enough for a mature core to assemble. We propose that inhibitors like PF-46396 bind to the Gag lattice where they deny the protease access to the CA-SP1 cleavage site and prevent the release of CA. CA5 particles, which exhibit no cleavage at the CA-SP1 site, have spheroidal shells with relatively thin walls. It appears that this lattice progresses displacively toward a mature-like state but produces neither conical cores nor infectious virions. These observations support the disassembly-reassembly pathway for core formation.


Subject(s)
Capsid/chemistry , Capsid/metabolism , HIV-1/metabolism , Rous sarcoma virus/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Gene Products, gag/chemistry , Gene Products, gag/genetics , Gene Products, gag/metabolism , HIV-1/chemistry , HIV-1/genetics , Humans , Models, Molecular , Mutation , Rous sarcoma virus/chemistry , Rous sarcoma virus/genetics , gag Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism
13.
J Biotechnol ; 165(1): 69-75, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23511652

ABSTRACT

Virus-like particles (VLPs) displaying antigen have been increasingly recognized as a potential vaccine in the livestock industry. In this study, Neospora caninum surface protein related sequence (NcSRS)2 was displayed on the surface of Rous sarcoma virus group-antigen protein (RSV-gag) VLPs. Two types of Bombyx mori nucleopolyhedrovirus (BmNPV) bacmids, encoding RSV-gag and NcSRS2 genes, were co-injected into silkworm larvae to produce VLPs-NcSRS2. At 7 days post-injection, VLPs-NcSRS2 were collected from hemolymph and purified. The antigenicity of the purified protein was confirmed by enzyme-linked immunosorbent assay (ELISA) using neosporosis-positive bovine serum. ELISA revealed that ~0.16µg rNcSRS2 was displayed per 1µg VLPs-NcSRS2. To develop an antibody specific for VLPs-NcSRS2, purified VLPs-NcSRS2 were used to immunize mice in a three-dose regimen without adjuvant and the production of antibodies was confirmed in serum samples. By using a silkworm expression system, we demonstrated the display, expression and immunization of neosporosis-targeting membrane proteins, which are vaccine candidates for neosporosis.


Subject(s)
Antigens, Protozoan/genetics , Antigens, Surface/genetics , Coccidiosis/genetics , Gene Products, gag/genetics , Protozoan Proteins/genetics , Vaccines, Virus-Like Particle/genetics , Animals , Antigens, Protozoan/metabolism , Antigens, Surface/metabolism , Bombyx/genetics , Cattle , Coccidiosis/immunology , Coccidiosis/prevention & control , Coccidiosis/veterinary , Gene Products, gag/metabolism , Neospora/genetics , Neospora/metabolism , Neospora/virology , Protozoan Proteins/metabolism , Rous sarcoma virus/genetics , Rous sarcoma virus/metabolism , Vaccines, Virus-Like Particle/biosynthesis , Vaccines, Virus-Like Particle/immunology
14.
Virus Res ; 171(2): 304-18, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23036987

ABSTRACT

The assembly and release of retrovirus particles from the cell membrane is directed by the Gag polyprotein. The Gag protein of Rous sarcoma virus (RSV) traffics through the nucleus prior to plasma membrane localization. We previously reported that nuclear localization of RSV Gag is linked to efficient packaging of viral genomic RNA, however the intranuclear activities of RSV Gag are not well understood. To gain insight into the properties of the RSV Gag protein within the nucleus, we examined the subnuclear localization and dynamic trafficking of RSV Gag. Restriction of RSV Gag to the nucleus by mutating its nuclear export signal (NES) in the p10 domain or interfering with CRM1-mediated nuclear export of Gag by leptomycin B (LMB) treatment led to the accumulation of Gag in nucleoli and discrete nucleoplasmic foci. Retention of RSV Gag in nucleoli was reduced with cis-expression of the 5' untranslated RU5 region of the viral RNA genome, suggesting the psi (Ψ) packaging signal may alter the subnuclear localization of Gag. Fluorescence recovery after photobleaching (FRAP) demonstrated that the nucleolar fraction of Gag was highly mobile, indicating that there was rapid exchange with Gag proteins in the nucleoplasm. RSV Gag is targeted to nucleoli by a nucleolar localization signal (NoLS) in the NC domain, and similarly, the human immunodeficiency virus type 1 (HIV-1) NC protein also contains an NoLS consisting of basic residues. Interestingly, co-expression of HIV-1 NC or Rev with HIV-1 Gag resulted in accumulation of Gag in nucleoli. Moreover, a subpopulation of HIV-1 Gag was detected in the nucleoli of HeLa cells stably expressing the entire HIV-1 genome in a Rev-dependent fashion. These findings suggest that the RSV and HIV-1 Gag proteins undergo nucleolar trafficking in the setting of viral infection.


Subject(s)
Cell Nucleolus/virology , Gene Products, gag/metabolism , HIV Infections/virology , HIV-1/metabolism , Rous sarcoma virus/metabolism , Sarcoma, Avian/virology , gag Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Nucleolus/metabolism , Gene Expression Regulation, Viral , Gene Products, gag/chemistry , Gene Products, gag/genetics , HIV Infections/metabolism , HIV-1/chemistry , HIV-1/genetics , Humans , Mice , Molecular Sequence Data , Nuclear Localization Signals , Protein Transport , Quail , Rous sarcoma virus/chemistry , Rous sarcoma virus/genetics , Sarcoma, Avian/metabolism , Sequence Alignment , gag Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/genetics
15.
Proteins ; 81(2): 316-25, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23011855

ABSTRACT

An infective retrovirus requires a mature capsid shell around the viral replication complex. This shell is formed by about 1500 capsid protein monomers, organized into hexamer and pentamer rings that are linked to each other by the dimerization of the C-terminal domain (CTD). The major homology region (MHR), the most highly conserved protein sequence across retroviral genomes, is part of the CTD. Several mutations in the MHR appear to block infectivity by preventing capsid formation. Suppressor mutations have been identified that are distant in sequence and structure from the MHR and restore capsid formation. The effects of two lethal and two suppressor mutations on the stability and function of the CTD were examined. No correlation with infectivity was found for the stability of the lethal mutations (D155Y-CTD, F167Y-CTD) and suppressor mutations (R185W-CTD, I190V-CTD). The stabilities of three double mutant proteins (D155Y/R185W-CTD, F167Y/R185W-CTD, and F167Y/I190V-CTD) were additive. However, the dimerization affinity of the mutant proteins correlated strongly with biological function. The CTD proteins with lethal mutations did not dimerize, while those with suppressor mutations had greater dimerization affinity than WT-CTD. The suppressor mutations were able to partially correct the dimerization defect caused by the lethal MHR mutations in double mutant proteins. Despite their dramatic effects on dimerization, none of these residues participate directly in the proposed dimerization interface in a mature capsid. These findings suggest that the conserved sequence of the MHR has critical roles in the conformation(s) of the CTD that are required for dimerization and correct capsid maturation.


Subject(s)
Capsid Proteins/genetics , Mutant Proteins/genetics , Mutation , Rous sarcoma virus/genetics , Capsid/metabolism , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Multimerization , Protein Structure, Tertiary , Rous sarcoma virus/metabolism , Sequence Homology, Amino Acid
16.
J Virol Methods ; 177(2): 147-52, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21816175

ABSTRACT

Rous sarcoma virus group antigen protein-based virus-like particles (VLPs) are well known for their structural integrity and ease of handling. VLPs play an important role in drug delivery systems because they can be manipulated with ease. In this study, a new method was established for expressing Rous sarcoma virus group antigen protein based VLPs in silkworm larvae and establishing stably expressing insect cell lines. These VLPs have been isolated by ultracentrifugation using a sucrose step gradient of 10-60% (v/v), and their spherical structure has been confirmed using transmission electron microscopy (TEM). The spherical morphology is similar in both the silkworm larvae and in stably expressing cell lines. Silkworm larvae are better suited for producing Rous sarcoma virus group antigen protein-based VLPs on a large scale; yields from silkworm larvae were approximately 8.2-fold higher than yields from stable cell lines. These VLPs provide a new method for large-scale application in vaccine development and drug delivery systems.


Subject(s)
Bombyx/metabolism , Gene Products, gag/metabolism , Genome, Insect , Rous sarcoma virus/metabolism , Animals , Antigens, Viral/metabolism , Blotting, Western , Bombyx/genetics , Cell Line , Genetic Vectors/genetics , Genetic Vectors/metabolism , Hemolymph/metabolism , Larva/genetics , Larva/metabolism , Microscopy, Electron, Transmission , Rous sarcoma virus/genetics , Ultracentrifugation , Viral Proteins/genetics , Viral Proteins/metabolism
17.
J Biotechnol ; 155(2): 185-92, 2011 Sep 10.
Article in English | MEDLINE | ID: mdl-21794838

ABSTRACT

Two types of Rous sarcoma virus (RSV) group-antigen protein (Gag) virus like particles (VLPs), full-length Gag (Gag701) and RSV protease domain (PR)-deleted mutant (Gag577) were expressed in silkworm larvae. Gag577 was secreted into hemolymph efficiently using wild type bacmid (WT), cysteine protease-deficient bacmid (CP(-)), cysteine protease and chitinase-deficient bacmid (CP(-)Chi(-)) bacmids, but comparatively Gag701 secretion levels were low. VLPs were purified on 10-60% (v/v) sucrose density gradient by ultracentrifugation and their structures confirmed under electron microscope. When hPRR and RSV Gag577 were co-expressed in silkworm larvae, human prorenin receptor (hPRR) was displayed on the surface of RSV VLPs, which was detected by Western blotting and immunoelectron microscopy. Moreover, binding of hPRR localized on the surface of VLPs to human prorenin was confirmed by ELISA. These results indicate that active hPRR was displayed on the surface of RSV VLPs, which can be utilized for drug discovery of hPRR blockers to prevent nephropathy. Moreover, this transmembrane protein display system using RSV Gag in silkworm larvae is applicable to expression of intact transmembrane proteins and binding assay of transmembrane proteins to its ligands, especially for transmembrane proteins which cannot be purified from membrane fractions in active states.


Subject(s)
Bombyx , Drug Discovery/methods , Gene Products, gag/metabolism , Receptors, Cell Surface/metabolism , Rous sarcoma virus/metabolism , Virion/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Humans , Larva/metabolism , Larva/virology , Microscopy, Immunoelectron , Ultracentrifugation , Virion/isolation & purification , Prorenin Receptor
18.
Proc Natl Acad Sci U S A ; 107(20): 9358-63, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20435918

ABSTRACT

Retroviral Gag polyproteins coopt host factors to traffic from cytosolic ribosomes to the plasma membrane, where virions are released. Before membrane transport, the multidomain Gag protein of Rous sarcoma virus (RSV) undergoes importin-mediated nuclear import and CRM1-dependent nuclear export, an intrinsic step in the assembly pathway. Transient nuclear trafficking of Gag is required for efficient viral RNA (vRNA) encapsidation, suggesting that Gag:vRNA binding might occur in the nucleus. Here, we show that Gag is imported into the nucleus through direct interactions of the Gag NC domain with importin-alpha (imp-alpha) and the MA domain with importin-11 (imp-11). The vRNA packaging signal, known as psi, inhibited imp-alpha binding to Gag, indicating that the NC domain does not bind to imp-alpha and vRNA simultaneously. Unexpectedly, vRNA binding also prevented the association of imp-11 with both the MA domain alone and with Gag, suggesting that the MA domain may bind to the vRNA genome. In contrast, direct binding of Gag to the nuclear export factor CRM1, via the CRM1-RanGTP heterodimer, was stimulated by psiRNA. These findings suggest a model whereby the genomic vRNA serves as a switch to regulate the ordered association of host import/export factors that mediate Gag nucleocytoplasmic trafficking for virion assembly. The Gag:vRNA interaction appears to serve multiple critical roles in assembly: specific selection of the vRNA genome for packaging, stimulating the formation of Gag dimers, and triggering export of viral ribonucleoprotein complexes from the nucleus.


Subject(s)
Cell Nucleus/metabolism , Gene Products, gag/metabolism , Karyopherins/metabolism , RNA, Viral/metabolism , Rous sarcoma virus/metabolism , Virus Assembly/physiology , Active Transport, Cell Nucleus/physiology , Animals , Blotting, Western , Cell Line , Immunoprecipitation , Microscopy, Confocal , Models, Biological , Quail , Ribonucleoproteins/metabolism , Rous sarcoma virus/genetics , Virus Assembly/genetics
19.
Biochemistry ; 49(19): 4006-17, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20387899

ABSTRACT

Assembly of retrovirus particles is promoted by interaction of the Gag polyprotein with RNA. Nonspecific RNA association with the nucleocapsid domain (NC) of Gag induces the dimerization of Gag through protein-protein contacts in the capsid domain (CA), followed by higher order assembly to form the immature virus particle. NMR relaxation studies were conducted to investigate the initial steps of Rous sarcoma virus (RSV) assembly by examining the association with nucleic acid of a fragment of Gag comprising the C-terminal domain of CA (CTD) postulated to mediate Gag dimerization, the spacer region between CA and NC (SP), and NC. This fragment, CTD-SP-NC (residues 394-577), spans the critical SP region and allows assessment of this key Gag-nucleic acid interaction in the context of the Gag polyprotein rather than the isolated domains. Main-chain amide relaxation of CTD-SP-NC was measured in the absence and presence of (GT)(4), an 8-mer DNA oligonucleotide that binds tightly to the polyprotein but is too short to promote Gag dimerization. The results show that the CTD and NC domains tumble independently. In contrast, the two zinc finger domains within NC are rotationally coupled in both the unbound and bound states, even though only the first zinc finger appears to make direct contact with (GT)(4). In addition, the NMR data indicate that SP and flanking residues undergo a conformational exchange process that is slowed in the presence of (GT)(4). This region around SP where relaxation is strongly affected by (GT)(4) binding is nearly identical to the assembly domain defined previously by mutagenesis studies. Other changes in relaxation induced by (GT)(4) implicate conformational perturbations of helices 1 and 4 in CTD. On the basis of the combined data, we propose a model for the promotion of Gag dimerization by RNA association in which NC-RNA binding disrupts an assembly inhibitory, intramolecular interaction involving SP and CTD. Disruption of this intramolecular interaction is proposed to enhance the accessibility of the Gag dimer contact surface and release the assembly domain to promote intermolecular oligomerization.


Subject(s)
Gene Products, gag/chemistry , Gene Products, gag/metabolism , RNA, Viral/chemistry , Rous sarcoma virus/metabolism , Base Sequence , Binding Sites , Gene Products, gag/antagonists & inhibitors , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Nucleocapsid/chemistry , Nucleocapsid/metabolism , Protein Structure, Secondary , RNA, Viral/metabolism
20.
Structure ; 17(5): 737-48, 2009 May 13.
Article in English | MEDLINE | ID: mdl-19446529

ABSTRACT

In mature retroviral particles, the capsid protein (CA) forms a shell encasing the viral replication complex. Human immunodeficiency virus (HIV) CA dimerizes in solution, through its C-terminal domain (CTD), and this interaction is important for capsid assembly. In contrast, other retroviral capsid proteins, including that of Rous sarcoma virus (RSV), do not dimerize with measurable affinity. Here we show, using X-ray crystallography and other biophysical methods, that acidification causes RSV CA to dimerize in a fashion analogous to HIV CA, and that this drives capsid assembly in vitro. A pair of aspartic acid residues, located within the CTD dimer interface, explains why dimerization is linked to proton binding. Our results show that despite overarching structural similarities, the intermolecular forces responsible for forming and stabilizing the retroviral capsid differ markedly across retroviral genera. Our data further suggest that proton binding may regulate RSV capsid assembly, or modulate stability of the assembled capsid.


Subject(s)
Capsid Proteins/chemistry , Capsid/metabolism , Protons , Rous sarcoma virus/metabolism , Viral Proteins/chemistry , Capsid/chemistry , Capsid Proteins/metabolism , Humans , Models, Molecular , Molecular Conformation , Protein Multimerization , Protein Structure, Tertiary , Viral Proteins/metabolism , Virus Assembly
SELECTION OF CITATIONS
SEARCH DETAIL
...