38 research outputs found

    Analysis of Prototype Foamy Virus particle-host cell interaction with autofluorescent retroviral particles

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>The foamy virus (FV) replication cycle displays several unique features, which set them apart from orthoretroviruses. First, like other B/D type orthoretroviruses, FV capsids preassemble at the centrosome, but more similar to hepadnaviruses, FV budding is strictly dependent on cognate viral glycoprotein coexpression. Second, the unusually broad host range of FV is thought to be due to use of a very common entry receptor present on host cell plasma membranes, because all cell lines tested in vitro so far are permissive.</p> <p>Results</p> <p>In order to take advantage of modern fluorescent microscopy techniques to study FV replication, we have created FV Gag proteins bearing a variety of protein tags and evaluated these for their ability to support various steps of FV replication. Addition of even small N-terminal HA-tags to FV Gag severely impaired FV particle release. For example, release was completely abrogated by an N-terminal autofluorescent protein (AFP) fusion, despite apparently normal intracellular capsid assembly. In contrast, C-terminal Gag-tags had only minor effects on particle assembly, egress and particle morphogenesis. The infectivity of C-terminal capsid-tagged FV vector particles was reduced up to 100-fold in comparison to wild type; however, infectivity was rescued by coexpression of wild type Gag and assembly of mixed particles. Specific dose-dependent binding of fluorescent FV particles to target cells was demonstrated in an Env-dependent manner, but not binding to target cell-extracted- or synthetic- lipids. Screening of target cells of various origins resulted in the identification of two cell lines, a human erythroid precursor- and a zebrafish- cell line, resistant to FV Env-mediated FV- and HIV-vector transduction.</p> <p>Conclusions</p> <p>We have established functional, autofluorescent foamy viral particles as a valuable new tool to study FV - host cell interactions using modern fluorescent imaging techniques. Furthermore, we succeeded for the first time in identifying two cell lines resistant to Prototype Foamy Virus Env-mediated gene transfer. Interestingly, both cell lines still displayed FV Env-dependent attachment of fluorescent retroviral particles, implying a post-binding block potentially due to lack of putative FV entry cofactors. These cell lines might ultimately lead to the identification of the currently unknown ubiquitous cellular entry receptor(s) of FVs.</p

    The Foamy Virus Gag Proteins: What Makes Them Different?

    Get PDF
    Gag proteins play an important role in many stages of the retroviral replication cycle. They orchestrate viral assembly, interact with numerous host cell proteins, engage in regulation of viral gene expression, and provide the main driving force for virus intracellular trafficking and budding. Foamy Viruses (FV), also known as spumaviruses, display a number of unique features among retroviruses. Many of these features can be attributed to their Gag proteins. FV Gag proteins lack characteristic orthoretroviral domains like membrane-binding domains (M domains), the major homology region (MHR), and the hallmark Cys-His motifs. In contrast, they contain several distinct domains such as the essential Gag-Env interaction domain and the glycine and arginine rich boxes (GR boxes). Furthermore, FV Gag only undergoes limited maturation and follows an unusual pathway for nuclear translocation. This review summarizes the known FV Gag domains and motifs and their functions. In particular, it provides an overview of the unique structural and functional properties that distinguish FV Gag proteins from orthoretroviral Gag proteins

    Novel Functions of Prototype Foamy Virus Gag Glycine- Arginine-Rich Boxes in Reverse Transcription and Particle Morphogenesis▿

    No full text
    Prototype foamy virus (PFV) Gag lacks the characteristic orthoretroviral Cys-His motifs that are essential for various steps of the orthoretroviral replication cycle, such as RNA packaging, reverse transcription, infectivity, integration, and viral assembly. Instead, it contains three glycine-arginine-rich boxes (GR boxes) in its C terminus that putatively represent a functional equivalent. We used a four-plasmid replication-deficient PFV vector system, with uncoupled RNA genome packaging and structural protein translation, to analyze the effects of deletion and various substitution mutations within each GR box on particle release, particle-associated protein composition, RNA packaging, DNA content, infectivity, particle morphology, and intracellular localization. The degree of viral particle release by all mutants was similar to that of the wild type. Only minimal effects on Pol encapsidation, exogenous reverse transcriptase (RT) activity, and genomic viral RNA packaging were observed. In contrast, particle-associated DNA content and infectivity were drastically reduced for all deletion mutants and were undetectable for all alanine substitution mutants. Furthermore, GR box I mutants had significant changes in particle morphology, and GR box II mutants lacked the typical nuclear localization pattern of PFV Gag. Finally, it could be shown that GR boxes I and III, but not GR box II, can functionally complement each other. It therefore appears that, similar to the orthoretroviral Cys-His motifs, the PFV Gag GR boxes are important for RNA encapsidation, genome reverse transcription, and virion infectivity as well as for particle morphogenesis

    The cooperative function of arginine residues in the Prototype Foamy Virus Gag C-terminus mediates viral and cellular RNA encapsidation.

    Get PDF
    International audienceBackgroundOne 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.ResultsHere 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 PFV capsids 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).ConclusionsTaken 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

    Nuclear translocation of Cyclin B1 marks the restriction point for terminal cell cycle exit in G2 phase

    No full text
    <div><p>Upon DNA damage, cell cycle progression is temporally blocked to avoid propagation of mutations. While transformed cells largely maintain the competence to recover from a cell cycle arrest, untransformed cells past the G1/S transition lose mitotic inducers, and thus the ability to resume cell division. This permanent cell cycle exit depends on p21, p53, and APC/C<sup>Cdh1</sup>. However, when and how permanent cell cycle exit occurs remains unclear. Here, we have investigated the cell cycle response to DNA damage in single cells that express Cyclin B1 fused to eYFP at the endogenous locus. We find that upon DNA damage Cyclin B1-eYFP continues to accumulate up to a threshold level, which is reached only in G2 phase. Above this threshold, a p21 and p53-dependent nuclear translocation required for APC/C<sup>Cdh1</sup>-mediated Cyclin B1-eYFP degradation is initiated. Thus, cell cycle exit is decoupled from activation of the DNA damage response in a manner that correlates to Cyclin B1 levels, suggesting that G2 activities directly feed into the decision for cell cycle exit. Once Cyclin B1-eYFP nuclear translocation occurs, checkpoint inhibition can no longer promote mitotic entry or re-expression of mitotic inducers, suggesting that nuclear translocation of Cyclin B1 marks the restriction point for permanent cell cycle exit in G2 phase.</p></div

    Assessing kinetics from fixed cells reveals activation of the mitotic entry network at the S/G2 transition

    Get PDF
    The ultimate aim of the cell cycle is to create an identical daughter cell. Therefore, correct progression through the different phases of the cell cycle is crucial to ensure faithful cell division. Successful execution of the different processes in the cell cycle is achieved by the coordinated action of a complex network of protein kinases and phosphatases at the centre of which stand Cyclin-Cdk complexes. Human cells possess a variety of cyclins and Cdks, which form complexes that regulate cell cycle transitions. In an unperturbed cell cycle, preparing a cell for mitosis requires faithful DNA replication and reorganisation of the cell’s structures and organelles. In this scenario, cells initiate successive waves of Cdk activity that orchestrate the timely and spatially controlled phosphorylation of a multitude of targets. In contrast, upon DNA damage cells must halt cell cycle progression in order to prevent mitotic entry of damaged cells and subsequently avoid potential propagation of mutations. Strict control of Cyclin-Cdk complexes is, therefore, essential both for correct cell division and to maintain genome integrity. However, the exact mechanisms underlying the activation of Cyclin- Cdk complexes in these different scenarios remain largely unknown. In this thesis, I have investigated several aspects of the regulation of Cdk activity both in the unperturbed cell cycle and during a DNA damage response. To address Cdk activity in the unperturbed cell cycle we established a novel quantitative immunofluorescence method and assessed the dynamics of cyclin accumulation and Cdk target phosphorylation in the unperturbed cell cycle. We found that the mitotic entry network first becomes activated at the S/G2 transition. This finding shifts the classical view of an abrupt Cdk activation at mitotic entry to an earlier and more gradual activation. Furthermore, it provides a potential link between S phase and mitosis, suggesting the existence of a mechanism that maintains pro-mitotic activities under a certain threshold until DNA replication is completed (Paper I). Interestingly, in parallel to an increase of pro-mitotic activities at the S/G2 transition, we observed a change in the localisation of Cyclin A2. Using genome-edited cell lines that express endogenous Cyclin A2-eYFP we were able to determine the cell cycle-dependent localisation of Cyclin A2 to the cytoplasm. Interestingly, despite coinciding with an increase of Cdk activity in the cell cycle we found that the cytoplasmic accumulation Cyclin A2 is modulated by p21 and the presence rather than activity of Cdk1. These findings suggest that complex formation and interaction with Cdk inhibitor proteins (CKI) might regulate Cyclin A2 localisation throughout the cell cycle (Paper IV). Despite not having uncovered a role for cytoplasmic Cyclin A2, we hypothesise that the cell cycle-dependent localisation of cyclins may be an important step to regulate Cdk activity. In order to understand how cells modulate Cdk activity upon DNA damage we made use of endogenously tagged cell lines expressing Cyclin B1-eYFP. We found that upon DNA damage cells continue to accumulate Cyclin B1 until reaching levels that are normally present in G2 phase. At this point, cells translocate Cyclin B1 to the nucleus in a p21 and p53- dependent manner where it is degraded by APC/CCdh1. We identified nuclear translocation and degradation of Cyclin B1 as a restriction point in the cell cycle when cells irreversibly exit the cell cycle and become senescent (Paper II). Senescence is regarded as an early barrier for tumorigenesis as it prevents the propagation of cells with damaged DNA. Our findings in Paper II suggested a link between mitotic inducers and the induction of senescence; therefore we decided to investigate the role of Cdk activity in terminal cell cycle exit. We found that upon DNA damage cells preserve low levels of Cdk activity to ensure that damaged cells continue to progress through the cell cycle until they reach a point where they can be forced into senescence. In this context, we found that Cdk activity induces p21 expression in a p53-independent manner to promote nuclear translocation and degradation of Cyclin B1 and other mitotic inducers (Paper III). Altogether, the data presented in this thesis points towards the existence of a link between the mitotic entry network and the DNA damage response to modulate the activity of Cyclin-Cdk complexes in time and space to trigger ensure correct progression to mitosis or, when needed, to trigger senescence
    corecore