Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 187(16): 4213-4230.e19, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39013471

ABSTRACT

Foamy viruses (FVs) are an ancient lineage of retroviruses, with an evolutionary history spanning over 450 million years. Vector systems based on Prototype Foamy Virus (PFV) are promising candidates for gene and oncolytic therapies. Structural studies of PFV contribute to the understanding of the mechanisms of FV replication, cell entry and infection, and retroviral evolution. Here we combine cryoEM and cryoET to determine high-resolution in situ structures of the PFV icosahedral capsid (CA) and envelope glycoprotein (Env), including its type III transmembrane anchor and membrane-proximal external region (MPER), and show how they are organized in an integrated structure of assembled PFV particles. The atomic models reveal an ancient retroviral capsid architecture and an unexpected relationship between Env and other class 1 fusion proteins of the Mononegavirales. Our results represent the de novo structure determination of an assembled retrovirus particle.


Subject(s)
Cryoelectron Microscopy , Spumavirus , Virus Assembly , Virus Internalization , Spumavirus/genetics , Capsid/metabolism , Capsid/chemistry , Capsid/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Capsid Proteins/genetics , Humans , Evolution, Molecular , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Models, Molecular
2.
Horm Metab Res ; 56(4): 308-317, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37793428

ABSTRACT

The emergence of SARS-CoV 2 caused the COVID-19 pandemic, resulting in numerous global infections and deaths. In particular, people with metabolic diseases display an increased risk of severe COVID 19 and a fatal outcome. Treatment options for severe cases are limited, and the appearance of new virus variants complicates the development of novel therapies. To better manage viral infections like COVID 19, new therapeutic approaches are needed. Marine sponges offer a natural and renewable source of unique bioactive agents. These sponges produce secondary metabolites with various effects, including anti-viral, anti-inflammatory, and anti-tumorigenic properties. In the current study, we investigated the effect of five different marine sponge-derived secondary metabolites (four bromotyrosines and one sesquiterpenoid hydroquinone). Two of these, Avarol and Acetyl-dibromoverongiaquinol reduced the expression of ACE2, the main receptor for SARS-CoV 2, and the alternative receptor NRP1. Moreover, these substances derived from sponges demonstrated the ability to diminish the virus titer in SARS-CoV 2-infected cells, especially concerning the Omicron lineage. However, the reduction was not substantial enough to expect a significant impact on infected humans. Consequently, the investigated sponge-derived secondary metabolites are not likely to be effective to treat COVID 19 as a stand-alone therapy.


Subject(s)
COVID-19 , Porifera , Animals , Humans , SARS-CoV-2 , Pandemics
3.
J Gen Virol ; 102(12)2021 12.
Article in English | MEDLINE | ID: mdl-34939563

ABSTRACT

Viruses in the family Retroviridae are found in a wide variety of vertebrate hosts. Enveloped virions are 80-100 nm in diameter with an inner core containing the viral genome and replicative enzymes. Core morphology is often characteristic for viruses within the same genus. Replication involves reverse transcription and integration into host cell DNA, resulting in a provirus. Integration into germline cells can result in a heritable provirus known as an endogenous retrovirus. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Retroviridae, which is available at ictv.global/report/retroviridae.


Subject(s)
DNA Viruses/classification , Retroviridae/classification , Animals , DNA Viruses/genetics , DNA Viruses/physiology , DNA Viruses/ultrastructure , Genome, Viral , Host Specificity , Retroviridae/genetics , Retroviridae/physiology , Retroviridae/ultrastructure , Vertebrates/virology , Virion/ultrastructure , Virus Replication
4.
Nature ; 523(7560): 366-9, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26061770

ABSTRACT

Retroviral integration is catalysed by a tetramer of integrase (IN) assembled on viral DNA ends in a stable complex, known as the intasome. How the intasome interfaces with chromosomal DNA, which exists in the form of nucleosomal arrays, is currently unknown. Here we show that the prototype foamy virus (PFV) intasome is proficient at stable capture of nucleosomes as targets for integration. Single-particle cryo-electron microscopy reveals a multivalent intasome-nucleosome interface involving both gyres of nucleosomal DNA and one H2A-H2B heterodimer. While the histone octamer remains intact, the DNA is lifted from the surface of the H2A-H2B heterodimer to allow integration at strongly preferred superhelix location ±3.5 positions. Amino acid substitutions disrupting these contacts impinge on the ability of the intasome to engage nucleosomes in vitro and redistribute viral integration sites on the genomic scale. Our findings elucidate the molecular basis for nucleosome capture by the viral DNA recombination machinery and the underlying nucleosome plasticity that allows integration.


Subject(s)
Nucleosomes/chemistry , Nucleosomes/virology , Spumavirus/metabolism , Virus Integration , Amino Acid Substitution , Binding Sites/genetics , Cryoelectron Microscopy , DNA/genetics , DNA/metabolism , DNA/ultrastructure , Genome/genetics , Histones/chemistry , Histones/metabolism , Histones/ultrastructure , Integrases/metabolism , Models, Molecular , Nucleosomes/genetics , Nucleosomes/ultrastructure , Protein Multimerization , Recombination, Genetic , Spumavirus/chemistry , Spumavirus/genetics , Spumavirus/ultrastructure
5.
PLoS Pathog ; 14(10): e1007293, 2018 10.
Article in English | MEDLINE | ID: mdl-30296302

ABSTRACT

Human diseases of zoonotic origin are a major public health problem. Simian foamy viruses (SFVs) are complex retroviruses which are currently spilling over to humans. Replication-competent SFVs persist over the lifetime of their human hosts, without spreading to secondary hosts, suggesting the presence of efficient immune control. Accordingly, we aimed to perform an in-depth characterization of neutralizing antibodies raised by humans infected with a zoonotic SFV. We quantified the neutralizing capacity of plasma samples from 58 SFV-infected hunters against primary zoonotic gorilla and chimpanzee SFV strains, and laboratory-adapted chimpanzee SFV. The genotype of the strain infecting each hunter was identified by direct sequencing of the env gene amplified from the buffy coat with genotype-specific primers. Foamy virus vector particles (FVV) enveloped by wild-type and chimeric gorilla SFV were used to map the envelope region targeted by antibodies. Here, we showed high titers of neutralizing antibodies in the plasma of most SFV-infected individuals. Neutralizing antibodies target the dimorphic portion of the envelope protein surface domain. Epitopes recognized by neutralizing antibodies have been conserved during the cospeciation of SFV with their nonhuman primate host. Greater neutralization breadth in plasma samples of SFV-infected humans was statistically associated with smaller SFV-related hematological changes. The neutralization patterns provide evidence for persistent expression of viral proteins and a high prevalence of coinfection. In conclusion, neutralizing antibodies raised against zoonotic SFV target immunodominant and conserved epitopes located in the receptor binding domain. These properties support their potential role in restricting the spread of SFV in the human population.


Subject(s)
Antibodies, Neutralizing/blood , Disease Vectors , Epitopes/immunology , Hominidae/immunology , Retroviridae Infections/transmission , Simian foamy virus/isolation & purification , Viral Envelope Proteins/immunology , Adult , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , Binding Sites , Gorilla gorilla/virology , Hominidae/blood , Hominidae/virology , Humans , Male , Middle Aged , Pan troglodytes/virology , Retroviridae Infections/virology
6.
Proc Natl Acad Sci U S A ; 114(21): 5509-5514, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28490494

ABSTRACT

The interactions between a retrovirus and host cell chromatin that underlie integration and provirus expression are poorly understood. The prototype foamy virus (PFV) structural protein GAG associates with chromosomes via a chromatin-binding sequence (CBS) located within its C-terminal region. Here, we show that the PFV CBS is essential and sufficient for a direct interaction with nucleosomes and present a crystal structure of the CBS bound to a mononucleosome. The CBS interacts with the histone octamer, engaging the H2A-H2B acidic patch in a manner similar to other acidic patch-binding proteins such as herpesvirus latency-associated nuclear antigen (LANA). Substitutions of the invariant arginine anchor residue in GAG result in global redistribution of PFV and macaque simian foamy virus (SFVmac) integration sites toward centromeres, dampening the resulting proviral expression without affecting the overall efficiency of integration. Our findings underscore the importance of retroviral structural proteins for integration site selection and the avoidance of genomic junkyards.


Subject(s)
Histones/metabolism , Nucleosomes/metabolism , Spumavirus/physiology , Virus Integration
7.
PLoS Pathog ; 12(7): e1005721, 2016 07.
Article in English | MEDLINE | ID: mdl-27399201

ABSTRACT

Foamy viruses (FV) belong to the genus Spumavirus, which forms a distinct lineage in the Retroviridae family. Although the infection in natural hosts and zoonotic transmission to humans is asymptomatic, FVs can replicate well in human cells making it an attractive gene therapy vector candidate. Here we present cryo-electron microscopy and (cryo-)electron tomography ultrastructural data on purified prototype FV (PFV) and PFV infected cells. Mature PFV particles have a distinct morphology with a capsid of constant dimension as well as a less ordered shell of density between the capsid and the membrane likely formed by the Gag N-terminal domain and the cytoplasmic part of the Env leader peptide gp18LP. The viral membrane contains trimeric Env glycoproteins partly arranged in interlocked hexagonal assemblies. In situ 3D reconstruction by subtomogram averaging of wild type Env and of a Env gp48TM- gp80SU cleavage site mutant showed a similar spike architecture as well as stabilization of the hexagonal lattice by clear connections between lower densities of neighboring trimers. Cryo-EM was employed to obtain a 9 Å resolution map of the glycoprotein in its pre-fusion state, which revealed extensive trimer interactions by the receptor binding subunit gp80SU at the top of the spike and three central helices derived from the fusion protein subunit gp48TM. The lower part of Env, presumably composed of interlaced parts of gp48TM, gp80SU and gp18LP anchors the spike at the membrane. We propose that the gp48TM density continues into three central transmembrane helices, which interact with three outer transmembrane helices derived from gp18LP. Our ultrastructural data and 9 Å resolution glycoprotein structure provide important new insights into the molecular architecture of PFV and its distinct evolutionary relationship with other members of the Retroviridae.


Subject(s)
Gene Products, env/ultrastructure , Glycoproteins/ultrastructure , Spumavirus/ultrastructure , Blotting, Western , Cell Line , Cryoelectron Microscopy , Humans , Image Processing, Computer-Assisted , Protein Conformation , Spumavirus/chemistry , Transfection
8.
PLoS Pathog ; 12(11): e1005981, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27829070

ABSTRACT

The Spumaretrovirinae, or foamy viruses (FVs) are complex retroviruses that infect many species of monkey and ape. Despite little sequence homology, FV and orthoretroviral Gag proteins perform equivalent functions, including genome packaging, virion assembly, trafficking and membrane targeting. However, there is a paucity of structural information for FVs and it is unclear how disparate FV and orthoretroviral Gag molecules share the same function. To probe the functional overlap of FV and orthoretroviral Gag we have determined the structure of a central region of Gag from the Prototype FV (PFV). The structure comprises two all α-helical domains NtDCEN and CtDCEN that although they have no sequence similarity, we show they share the same core fold as the N- (NtDCA) and C-terminal domains (CtDCA) of archetypal orthoretroviral capsid protein (CA). Moreover, structural comparisons with orthoretroviral CA align PFV NtDCEN and CtDCEN with NtDCA and CtDCA respectively. Further in vitro and functional virological assays reveal that residues making inter-domain NtDCEN-CtDCEN interactions are required for PFV capsid assembly and that intact capsid is required for PFV reverse transcription. These data provide the first information that relates the Gag proteins of Spuma and Orthoretrovirinae and suggests a common ancestor for both lineages containing an ancient CA fold.


Subject(s)
Capsid Proteins/genetics , Gene Products, gag/chemistry , Gene Products, gag/genetics , Spumavirus/genetics , Virus Assembly/physiology , Amino Acid Sequence , Animals , Blotting, Western , Capsid , Cell Line , Humans , Nuclear Magnetic Resonance, Biomolecular , Protein Conformation , Real-Time Polymerase Chain Reaction
9.
PLoS Pathog ; 12(8): e1005860, 2016 08.
Article in English | MEDLINE | ID: mdl-27579920

ABSTRACT

Unlike for other retroviruses, only a few host cell factors that aid the replication of foamy viruses (FVs) via interaction with viral structural components are known. Using a yeast-two-hybrid (Y2H) screen with prototype FV (PFV) Gag protein as bait we identified human polo-like kinase 2 (hPLK2), a member of cell cycle regulatory kinases, as a new interactor of PFV capsids. Further Y2H studies confirmed interaction of PFV Gag with several PLKs of both human and rat origin. A consensus Ser-Thr/Ser-Pro (S-T/S-P) motif in Gag, which is conserved among primate FVs and phosphorylated in PFV virions, was essential for recognition by PLKs. In the case of rat PLK2, functional kinase and polo-box domains were required for interaction with PFV Gag. Fluorescently-tagged PFV Gag, through its chromatin tethering function, selectively relocalized ectopically expressed eGFP-tagged PLK proteins to mitotic chromosomes in a Gag STP motif-dependent manner, confirming a specific and dominant nature of the Gag-PLK interaction in mammalian cells. The functional relevance of the Gag-PLK interaction was examined in the context of replication-competent FVs and single-round PFV vectors. Although STP motif mutated viruses displayed wild type (wt) particle release, RNA packaging and intra-particle reverse transcription, their replication capacity was decreased 3-fold in single-cycle infections, and up to 20-fold in spreading infections over an extended time period. Strikingly similar defects were observed when cells infected with single-round wt Gag PFV vectors were treated with a pan PLK inhibitor. Analysis of entry kinetics of the mutant viruses indicated a post-fusion defect resulting in delayed and reduced integration, which was accompanied with an enhanced preference to integrate into heterochromatin. We conclude that interaction between PFV Gag and cellular PLK proteins is important for early replication steps of PFV within host cells.


Subject(s)
Capsid/metabolism , Protein Serine-Threonine Kinases/metabolism , Retroviridae Infections/metabolism , Spumavirus/metabolism , Virus Integration/physiology , Amino Acid Motifs , Animals , Gene Products, gag/genetics , Gene Products, gag/metabolism , HeLa Cells , Humans , Mice , Phosphorylation/genetics , Protein Domains , Protein Serine-Threonine Kinases/genetics , Rats , Retroviridae Infections/genetics , Spumavirus/genetics
11.
J Immunol ; 194(7): 3201-12, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25740942

ABSTRACT

NK cells are emerging as new effectors for immunotherapy of cancer. In particular, the genetic engraftment of chimeric Ag receptors (CARs) in NK cells is a promising strategy to redirect NK cells to otherwise NK cell-resistant tumor cells. On the basis of DNAX-activation protein 12 (DAP12), a signaling adaptor molecule involved in signal transduction of activating NK cell receptors, we generated a new type of CAR targeting the prostate stem cell Ag (PSCA). We demonstrate in this article that this CAR, designated anti-PSCA-DAP12, consisting of DAP12 fused to the anti-PSCA single-chain Ab fragment scFv(AM1) confers improved cytotoxicity to the NK cell line YTS against PSCA-positive tumor cells when compared with a CAR containing the CD3ζ signaling chain. Further analyses revealed phosphorylation of the DAP12-associated ZAP-70 kinase and IFN-γ release of CAR-engineered cells after contact with PSCA-positive target cells. YTS cells modified with DAP12 alone or with a CAR bearing a phosphorylation-defective ITAM were not activated. Notably, infused YTS cells armed with anti-PSCA-DAP12 caused delayed tumor xenograft growth and resulted in complete tumor eradication in a significant fraction of treated mice. The feasibility of the DAP12-based CAR was further tested in human primary NK cells and confers specific cytotoxicity against KIR/HLA-matched PSCA-positive tumor cells, which was further enhanced by KIR-HLA mismatches. We conclude that NK cells engineered with DAP12-based CARs are a promising tool for adoptive tumor immunotherapy.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Membrane Proteins/genetics , Neoplasms/genetics , Neoplasms/immunology , Receptors, Natural Killer Cell/genetics , Recombinant Fusion Proteins , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , CD3 Complex/genetics , CD3 Complex/immunology , Cell Line , Cell Line, Tumor , Cytotoxicity, Immunologic , Disease Models, Animal , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Genetic Vectors/genetics , Humans , Immunophenotyping , Immunotherapy , Immunotherapy, Adoptive , Interferon-gamma/biosynthesis , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms/metabolism , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Phenotype , Phosphorylation , Xenograft Model Antitumor Assays , ZAP-70 Protein-Tyrosine Kinase/metabolism
12.
Mol Ther ; 24(7): 1227-36, 2016 08.
Article in English | MEDLINE | ID: mdl-27133965

ABSTRACT

Mesenchymal stromal cells (MSCs) hold great promise for regenerative medicine. Stable ex vivo gene transfer to MSCs could improve the outcome and scope of MSC therapy, but current vectors require multiple rounds of transduction, involve genotoxic viral promoters and/or the addition of cytotoxic cationic polymers in order to achieve efficient transduction. We describe a self-inactivating foamy virus vector (FVV), incorporating the simian macaque foamy virus envelope and using physiological promoters, which efficiently transduces murine MSCs (mMSCs) in a single-round. High and sustained expression of the transgene, whether GFP or the lysosomal enzyme, arylsulphatase A (ARSA), was achieved. Defining MSC characteristics (surface marker expression and differentiation potential), as well as long-term engraftment and distribution in the murine brain following intracerebroventricular delivery, are unaffected by FVV transduction. Similarly, greater than 95% of human MSCs (hMSCs) were stably transduced using the same vector, facilitating human application. This work describes the best stable gene transfer vector available for mMSCs and hMSCs.


Subject(s)
Gene Transfer Techniques , Genetic Vectors/genetics , Mesenchymal Stem Cells/metabolism , Spumavirus/genetics , Transduction, Genetic , Animals , Cell Line , Gene Expression , Gene Order , Humans , Mesenchymal Stem Cell Transplantation , Mice , Promoter Regions, Genetic , Transgenes
13.
J Immunol ; 192(9): 4379-85, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24706726

ABSTRACT

The proinflammatory enzyme caspase-1 plays an important role in the innate immune system and is involved in a variety of inflammatory conditions. Rare naturally occurring human variants of the caspase-1 gene (CASP1) lead to different protein expression and structure and to decreased or absent enzymatic activity. Paradoxically, a significant number of patients with such variants suffer from febrile episodes despite decreased IL-1ß production and secretion. In this study, we investigate how variant (pro)caspase-1 can possibly contribute to inflammation. In a transfection model, such variant procaspase-1 binds receptor interacting protein kinase 2 (RIP2) via Caspase activation and recruitment domain (CARD)/CARD interaction and thereby activates NF-κB, whereas wild-type procaspase-1 reduces intracellular RIP2 levels by enzymatic cleavage and release into the supernatant. We approach the protein interactions by coimmunoprecipitation and confocal microscopy and show that NF-κB activation is inhibited by anti-RIP2-short hairpin RNA and by the expression of a RIP2 CARD-only protein. In conclusion, variant procaspase-1 binds RIP2 and thereby activates NF-κB. This pathway could possibly contribute to proinflammatory signaling.


Subject(s)
Caspase 1/genetics , Fever/genetics , Inflammation/genetics , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Blotting, Western , Caspase 1/metabolism , Fever/enzymology , Fluorescent Antibody Technique , Gene Knockdown Techniques , Genetic Variation , HEK293 Cells , Humans , Immunoprecipitation , Inflammation/immunology , Inflammation/metabolism , Signal Transduction/physiology , Transduction, Genetic , Transfection
14.
PLoS Pathog ; 9(5): e1003376, 2013 May.
Article in English | MEDLINE | ID: mdl-23675305

ABSTRACT

The Spumaretrovirinae, or foamyviruses (FVs) are complex retroviruses that infect many species of monkey and ape. Although FV infection is apparently benign, trans-species zoonosis is commonplace and has resulted in the isolation of the Prototypic Foamy Virus (PFV) from human sources and the potential for germ-line transmission. Despite little sequence homology, FV and orthoretroviral Gag proteins perform equivalent functions, including genome packaging, virion assembly, trafficking and membrane targeting. In addition, PFV Gag interacts with the FV Envelope (Env) protein to facilitate budding of infectious particles. Presently, there is a paucity of structural information with regards FVs and it is unclear how disparate FV and orthoretroviral Gag molecules share the same function. Therefore, in order to probe the functional overlap of FV and orthoretroviral Gag and learn more about FV egress and replication we have undertaken a structural, biophysical and virological study of PFV-Gag. We present the crystal structure of a dimeric amino terminal domain from PFV, Gag-NtD, both free and in complex with the leader peptide of PFV Env. The structure comprises a head domain together with a coiled coil that forms the dimer interface and despite the shared function it is entirely unrelated to either the capsid or matrix of Gag from other retroviruses. Furthermore, we present structural, biochemical and virological data that reveal the molecular details of the essential Gag-Env interaction and in addition we also examine the specificity of Trim5α restriction of PFV. These data provide the first information with regards to FV structural proteins and suggest a model for convergent evolution of gag genes where structurally unrelated molecules have become functionally equivalent.


Subject(s)
Biological Evolution , Capsid/metabolism , Gene Products, gag/chemistry , Gene Products, gag/metabolism , Simian foamy virus/metabolism , Amino Acid Sequence , Capsid/chemistry , Cell Line , Gene Products, gag/genetics , Humans , Molecular Sequence Data , Protein Structure, Quaternary , Protein Structure, Tertiary , Simian foamy virus/chemistry , Simian foamy virus/genetics , Transfection
15.
Mol Ther ; 22(8): 1460-1471, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24814152

ABSTRACT

Vector systems based on different retroviruses are widely used to achieve stable integration and expression of transgenes. More recently, transient genetic manipulation systems were developed that are based on integration- or reverse transcription-deficient retroviruses. Lack of viral genome integration is desirable not only for reducing tumorigenic potential but also for applications requiring transient transgene expression such as reprogramming or genome editing. However, all existing transient retroviral vector systems rely on virus-encoded encapsidation sequences for the transfer of heterologous genetic material. We discovered that the transient transgene expression observed in target cells transduced by reverse transcriptase-deficient foamy virus (FV) vectors is the consequence of subgenomic RNA encapsidation into FV particles. Based on this initial observation, we describe here the establishment of FV vectors that enable the efficient transient expression of various transgenes by packaging, transfer, and de novo translation of nonviral RNAs both in vitro and in vivo. Transient transgene expression levels were comparable to integrase-deficient vectors but, unlike the latter, declined to background levels within a few days. Our results show that this new FV vector system provides a useful, novel tool for efficient transient genetic manipulation of target tissues by transfer of nonviral RNAs.


Subject(s)
Fibroblasts/virology , RNA/metabolism , Spumavirus/genetics , Transduction, Genetic , Animals , Cell Line, Tumor , Fibroblasts/cytology , Gene Transfer Techniques , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , In Vitro Techniques , Mice , RNA-Directed DNA Polymerase/metabolism , Spumavirus/metabolism , Transgenes
16.
Retrovirology ; 11: 87, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25292281

ABSTRACT

BACKGROUND: One unique feature of the foamy virus (FV) capsid protein Gag is the absence of Cys-His motifs, which in orthoretroviruses are irreplaceable for multitude functions including viral RNA genome recognition and packaging. Instead, FV Gag contains glycine-arginine-rich (GR) sequences at its C-terminus. In case of prototype FV (PFV) these are historically grouped in three boxes, which have been shown to play essential functions in genome reverse transcription, virion infectivity and particle morphogenesis. Additional functions for RNA packaging and Pol encapsidation were suggested, but have not been conclusively addressed. RESULTS: Here we show that released wild type PFV particles, like orthoretroviruses, contain various cellular RNAs in addition to viral genome. Unlike orthoretroviruses, the content of selected cellular RNAs in capsids of PFV vector particles was not altered by viral genome encapsidation. Deletion of individual GR boxes had only minor negative effects (2 to 4-fold) on viral and cellular RNA encapsidation over a wide range of cellular Gag to viral genome ratios examined. Only the concurrent deletion of all three PFV Gag GR boxes, or the substitution of multiple arginine residues residing in the C-terminal GR box region by alanine, abolished both viral and cellular RNA encapsidation (>50 to >3,000-fold reduced), independent of the viral production system used. Consequently, those mutants also lacked detectable amounts of encapsidated Pol and were non-infectious. In contrast, particle release was reduced to a much lower extent (3 to 20-fold). CONCLUSIONS: Taken together, our data provides the first identification of a full-length PFV Gag mutant devoid in genome packaging and the first report of cellular RNA encapsidation into PFV particles. Our results suggest that the cooperative action of C-terminal clustered positively charged residues, present in all FV Gag proteins, is the main viral protein determinant for viral and cellular RNA encapsidation. The viral genome independent efficiency of cellular RNA encapsidation suggests differential packaging mechanisms for both types of RNAs. Finally, this study indicates that analogous to orthoretroviruses, Gag - nucleic acid interactions are required for FV capsid assembly and efficient particle release.


Subject(s)
Arginine/metabolism , Gene Products, gag/metabolism , RNA/metabolism , Spumavirus/physiology , Virus Assembly , Amino Acid Substitution , Cell Line , Gene Products, gag/genetics , Humans , Mutation, Missense , Sequence Deletion , Spumavirus/genetics
17.
J Virol ; 87(6): 3163-76, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23283957

ABSTRACT

Foamy viruses (FVs) are unique among retroviruses in performing genome reverse transcription (RTr) late in replication, resulting in an infectious DNA genome, and also in their unusual Pol biosynthesis and encapsidation strategy. In addition, FVs display only very limited Gag and Pol processing by the viral protease (PR) during particle morphogenesis and disassembly, both thought to be crucial for viral infectivity. Here, we report the generation of functional prototype FV (PFV) particles from mature or partially processed viral capsid and enzymatic proteins with infectivity levels of up to 20% of the wild type. Analysis of protein and nucleic acid composition, as well as infectivity, of virions generated from different Gag and Pol combinations (including both expression-optimized and authentic PFV open reading frames [ORFs]) revealed that precursor processing of Gag, but not Pol, during particle assembly is essential for production of infectious virions. Surprisingly, when processed Gag (instead of Gag precursor) was provided together with PR-deficient Pol precursor during virus production, infectious, viral DNA-containing particles were obtained, even when different vector or proviral expression systems were used. Although virion infectivity was reduced to 0.5 to 2% relative to that of the respective parental constructs, this finding overturns the current dogma in the FV literature that viral PR activity is absolutely essential at some point during target cell entry. Furthermore, it demonstrates that viral PR-mediated Gag precursor processing during particle assembly initiates intraparticle RTr. Finally, it shows that reverse transcriptase (RT) and integrase are enzymatically active in the Pol precursor within the viral capsid, thus enabling productive host cell infection.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Reverse Transcription , Spumavirus/enzymology , Spumavirus/physiology , Virus Uncoating , Cell Line , Humans , Virion/chemistry , Virion/metabolism
19.
Cell Microbiol ; 15(2): 227-36, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23051660

ABSTRACT

Foamy viruses (FVs), a unique type of retroviruses, are characterized by several unusual features in their replication strategy. FVs, common to all non-human primates and several other species, display an extremely broad tropism in vitro. Basically, all mammalian cells and species examined, but also cells of amphibian or bird origin, are permissive to FV glycoprotein (Env)-mediated capsid release into the cytoplasm. The nature of the broadly expressed, and potentially evolutionary conserved, FV entry receptor molecule(s) is poorly characterized. Although recent data indicate that proteoglycans serve as an important factor for FV Env-mediated target cell attachment, additional uncharacterized molecules appear to be essential for the pH-dependent fusion of viral and cellular lipid membranes after endocytic uptake of virions. Furthermore, FVs show a very special assembly strategy. Unlike other retroviruses, the FV capsid precursor protein (Gag) undergoes only very limited proteolytic processing during assembly. This results in an immature morphology of capsids found in released FV virions. In addition, the FV Gag protein appears to lack a functional membrane-targeting signal. As a consequence, FVs utilize a specific interaction between capsid and cognate viral glycoprotein for initiation of thebudding process. Genetic fusion of heterologous targeting domains for plasma but not endosomal membranes to FV Gag enables glycoprotein-independent particle egress. However, this is at the expense of normal capsid morphogenesis and infectivity. The low-level Gag precursor processing and the requirement for a reversible, artificial Gag membrane association for effective pseudotyping of FV capsids by heterologous glycoproteins strongly suggest that FVs require a transient interaction of capsids with cellular membranes for viral replication. Under natural condition, this appears to be achieved by the lack of a membrane-targeting function of the FV Gag protein and the accomplishment of capsid membrane attachment through an unusual specific interaction with the cognate glycoprotein.


Subject(s)
Capsid/chemistry , Gene Products, gag/genetics , Spumavirus/chemistry , Virion/chemistry , Virus Assembly/physiology , Animals , Capsid/metabolism , Capsid/ultrastructure , Cell Membrane/chemistry , Cell Membrane/virology , Endocytosis , Gene Products, gag/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Membrane Fusion , Spumavirus/metabolism , Spumavirus/ultrastructure , Virion/metabolism , Virion/ultrastructure , Virus Internalization , Virus Replication
20.
J Immunol ; 189(6): 3249-59, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22875801

ABSTRACT

Prostate cancer is the most common noncutaneous malignancy in men. The prostate stem cell Ag (PSCA) is a promising target for immunotherapy of advanced disease. Based on a novel mAb directed to PSCA, we established and compared a series of murine and humanized anti-CD3-anti-PSCA single-chain bispecific Abs. Their capability to redirect T cells for killing of tumor cells was analyzed. During these studies, we identified a novel bispecific humanized Ab that efficiently retargets T cells to tumor cells in a strictly Ag-dependent manner and at femtomolar concentrations. T cell activation, cytokine release, and lysis of target cells depend on a cross-linkage of redirected T cells with tumor cells, whereas binding of the anti-CD3 domain alone does not lead to an activation or cytokine release. Interestingly, both CD8+ and CD4+ T cells are activated in parallel and can efficiently mediate the lysis of tumor cells. However, the onset of killing via CD4+ T cells is delayed. Furthermore, redirecting T cells via the novel humanized bispecific Abs results in a delay of tumor growth in xenografted nude mice.


Subject(s)
Antibodies, Bispecific/physiology , Antibodies, Monoclonal, Humanized/physiology , Antigens, Neoplasm/biosynthesis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/immunology , Stem Cells/immunology , Tumor Stem Cell Assay , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Cell Death/immunology , Epitopes, T-Lymphocyte/immunology , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/immunology , Humans , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Prostatic Neoplasms/pathology , Stem Cells/pathology , Tumor Stem Cell Assay/methods
SELECTION OF CITATIONS
SEARCH DETAIL