20 research outputs found

    Manipulation of the ubiquitin-proteasome system by HIV-1 : role of the accessory protein Vpr

    Get PDF
    Le virus de l’immunodĂ©ficience humaine de type 1 (VIH-1), l’agent Ă©tiologique du SIDA, est un rĂ©trovirus complexe arborant plusieurs protĂ©ines accessoires : Nef, Vif, Vpr, et Vpu. Celles-ci sont impliquĂ©es dans la modulation de la rĂ©plication virale, dans l’évasion immunitaire et dans la progression de la pathogenĂšse du SIDA. Dans ce contexte, il a Ă©tĂ© dĂ©montrĂ© que la protĂ©ine virale R (Vpr) induit un arrĂȘt de cycle cellulaire en phase G2. Le mĂ©canisme par lequel Vpr exerce cette fonction est l’activation, ATR (Ataxia telangiectasia and Rad3 related)-dĂ©pendante, du point de contrĂŽle de dommage Ă  l’ADN, mais les facteurs et mĂ©canismes molĂ©culaires directement impliquĂ©s dans cette activitĂ© demeurent inconnus. Afin d’identifier de nouveaux facteurs cellulaires interagissant avec Vpr, nous avons utilisĂ© une purification d’affinitĂ© en tandem (TAP) pour isoler des complexes protĂ©iques natifs contenant Vpr. Nous avons dĂ©couvert que Vpr s’associait avec CRL4A(VprBP), un complexe cellulaire d’E3 ubiquitine ligase, comprenant les protĂ©ines Cullin 4A, DDB1 (DNA damage-binding protein 1) et VprBP (Vpr-binding protein). Nos Ă©tudes ont mis en Ă©vidence que le recrutement de la E3 ligase par Vpr Ă©tait nĂ©cessaire mais non suffisant pour l’induction de l’arrĂȘt de cycle cellulaire en G2, suggĂ©rant ainsi que des Ă©vĂ©nements additionnels seraient impliquĂ©s dans ce processus. À cet Ă©gard, nous apportons des preuves directes que Vpr dĂ©tourne les fonctions de CRL4A(VprBP) pour induire la polyubiquitination de type K48 et la dĂ©gradation protĂ©osomale de protĂ©ines cellulaires encore inconnues. Ces Ă©vĂ©nements d’ubiquitination induits par Vpr ont Ă©tĂ© dĂ©montrĂ©s comme Ă©tant nĂ©cessaire Ă  l’activation d’ATR. Finalement, nous montrons que Vpr forme des foyers ancrĂ©s Ă  la chromatine co-localisant avec VprBP ainsi qu’avec des facteurs impliquĂ©s dans la rĂ©paration de l’ADN. La formation de ces foyers reprĂ©sente un Ă©vĂ©nement essentiel et prĂ©coce dans l’induction de l’arrĂȘt de cycle cellulaire en G2. Enfin, nous dĂ©montrons que Vpr est capable de recruter CRL4A(VprBP) au niveau de la chromatine et nous apportons des preuves indiquant que le substrat inconnu ciblĂ© par Vpr est une protĂ©ine associĂ©e Ă  la chromatine. Globalement, nos rĂ©sultats rĂ©vĂšlent certains des mĂ©nanismes par lesquels Vpr induit des perturbations du cycle cellulaire. En outre, cette Ă©tude contribue Ă  notre comprĂ©hension de la modulation du systĂšme ubiquitine-protĂ©asome par le VIH-1 et son implication fonctionnelle dans la manipulation de l’environnement cellulaire de l’hĂŽte.Human immunodeficiency virus 1 (HIV-1), the etiologic agent of AIDS, is a complex retrovirus with several accessory proteins. HIV-1 accessory proteins Nef, Vif, Vpr, and Vpu have been implicated in the modulation of viral replication, enhancement of viral fitness, immune evasion, and progression of AIDS pathogenesis. In that regard, viral protein R (Vpr) induces a cell cycle arrest in the G2 phase by activating the canonical ATR (Ataxia telangiectasia and Rad3 related)-mediated DNA damage checkpoint, but cellular factors and mechanisms directly engaged in this process remain unknown. To identify novel Vpr-interacting cellular factors, we used tandem affinity purification (TAP) to isolate native Vpr-containing complexes. We found that Vpr hijacks a cellular E3 ubiquitin ligase complex, CRL4A(VprBP), composed of Cullin 4A, DDB1 (DNA damage-binding protein 1) and VprBP (Vpr-binding protein). Moreover, we observed that recruitment of the E3 ligase by Vpr was necessary but not sufficient for the induction of G2 cell cycle arrest, suggesting that additional events are involved. In this context, we provide direct evidence that Vpr usurps the function of CRL4A(VprBP) to induce the K48-linked polyubiquitination and proteasomal degradation of as-yet-unknown cellular proteins. These ubiquitination events mediated by Vpr were necessary for the activation of ATR. Moreover, we show that Vpr forms chromatin-associated foci that co-localize with VprBP and DNA repair factors. Our data indicate that formation of these foci represent a critical early event in the induction of G2 arrest. Finally, we show that Vpr is able to recruit CRL4A(VprBP) on chromatin and we provide evidence that the unknown substrate targeted by Vpr is a chromatin-associated protein. Overall, our results reveal some of the mechanisms by which Vpr induces cell cycle perturbations. Furthermore, this study contributes to our understanding of the modulation of the ubiquitin-proteasome system by HIV-1 and its functional implication in the manipulation of the host cellular environment

    HIV-1 Vpr up-regulates expression of ligands for the activating NKG2D receptor and promotes NK cell-mediated killing

    Full text link
    HIV upregulates cell-surface expression of specific ligands for the activating NKG2D receptor, including ULBP-1, -2, -3, but not MICA or MICB, in infected cells both in vitro and in vivo. However, the viral factor(s) involved in NKG2D ligand expression still remains undefined. HIV-1 Vpr activates the DNA damage/stress-sensing ATR kinase and promotes G2 cell-cycle arrest, conditions known to upregulate NKG2D ligands. We report here that HIV-1 selectively induces cell-surface expression of ULBP-2 in primary CD4+ T-lymphocytes by a process that is Vpr-dependent. Importantly, Vpr enhanced the susceptibility of HIV-1-infected cells to NK cell-mediated killing. Strikingly, Vpr alone was sufficient to upregulate expression of all NKG2D ligands and thus promoted efficient NKG2D-dependent NK cell-mediated killing. Delivery of virion-associated Vpr via defective HIV-1 particles induced analogous biological effects in non-infected target cells, suggesting that Vpr may act similarly beyond infected cells. All these activities relied on Vpr ability to activate the ATR-mediated DNA damage/stress checkpoint. Overall, these results indicate that Vpr is a key determinant responsible for HIV-1-induced upregulation of NKG2D ligands and further suggest an immunomodulatory role for Vpr that may not only contribute to HIV-1-induced CD4+ T-lymphocyte depletion but may also take part in HIV-1-induced NK cell dysfunction.JR is recipient of a Frederick Banting and Charles Best scholarship from the Canadian Institutes of Health Research (CIHR) while JPB is recipient of a CIHR studentship. EAC holds the Canada Research Chair in Human Retrovirology. This work was supported by grants from CIHR and the Fonds de recherche en santé du Québec AIDS network to EAC

    HIV-1 Vpr-Mediated G2 Arrest Involves the DDB1-CUL4AVPRBP E3 Ubiquitin Ligase

    Get PDF
    Human immunodeficiency virus type 1 (HIV-1) viral protein R (Vpr) has been shown to cause G2 cell cycle arrest in human cells by inducing ATR-mediated inactivation of p34cdc2, but factors directly engaged in this process remain unknown. We used tandem affinity purification to isolate native Vpr complexes. We found that damaged DNA binding protein 1 (DDB1), viral protein R binding protein (VPRBP), and cullin 4A (CUL4A)—components of a CUL4A E3 ubiquitin ligase complex, DDB1-CUL4AVPRBP—were able to associate with Vpr. Depletion of VPRBP by small interfering RNA impaired Vpr-mediated induction of G2 arrest. Importantly, VPRBP knockdown alone did not affect normal cell cycle progression or activation of ATR checkpoints, suggesting that the involvement of VPRBP in G2 arrest was specific to Vpr. Moreover, leucine/isoleucine-rich domain Vpr mutants impaired in their ability to interact with VPRBP and DDB1 also produced strongly attenuated G2 arrest. In contrast, G2 arrest–defective C-terminal Vpr mutants were found to maintain their ability to associate with these proteins, suggesting that the interaction of Vpr with the DDB1-VPRBP complex is necessary but not sufficient to block cell cycle progression. Overall, these results point toward a model in which Vpr could act as a connector between the DDB1-CUL4AVPRBP E3 ubiquitin ligase complex and an unknown cellular factor whose proteolysis or modulation of activity through ubiquitination would activate ATR-mediated checkpoint signaling and induce G2 arrest

    RNA-binding protein CPEB1 remodels host and viral RNA landscapes.

    Get PDF
    Host and virus interactions occurring at the post-transcriptional level are critical for infection but remain poorly understood. Here, we performed comprehensive transcriptome-wide analyses revealing that human cytomegalovirus (HCMV) infection results in widespread alternative splicing (AS), shortening of 3' untranslated regions (3' UTRs) and lengthening of poly(A)-tails in host gene transcripts. We found that the host RNA-binding protein CPEB1 was highly induced after infection, and ectopic expression of CPEB1 in noninfected cells recapitulated infection-related post-transcriptional changes. CPEB1 was also required for poly(A)-tail lengthening of viral RNAs important for productive infection. Strikingly, depletion of CPEB1 reversed infection-related cytopathology and post-transcriptional changes, and decreased productive HCMV titers. Host RNA processing was also altered in herpes simplex virus-2 (HSV-2)-infected cells, thereby indicating that this phenomenon might be a common occurrence during herpesvirus infections. We anticipate that our work may serve as a starting point for therapeutic targeting of host RNA-binding proteins in herpesvirus infections

    Formation of Mobile Chromatin-Associated Nuclear Foci Containing HIV-1 Vpr and VPRBP Is Critical for the Induction of G2 Cell Cycle Arrest

    Get PDF
    HIV-1 Viral protein R (Vpr) induces a cell cycle arrest at the G2/M phase by activating the ATR DNA damage/stress checkpoint. Recently, we and several other groups showed that Vpr performs this activity by recruiting the DDB1-CUL4A (VPRBP) E3 ubiquitin ligase. While recruitment of this E3 ubiquitin ligase complex has been shown to be required for G2 arrest, the subcellular compartment where this complex forms and functionally acts is unknown. Herein, using immunofluorescence and confocal microscopy, we show that Vpr forms nuclear foci in several cell types including HeLa cells and primary CD4+ T-lymphocytes. These nuclear foci contain VPRBP and partially overlap with DNA repair foci components such as Îł-H2AX, 53BP1 and RPA32. While treatment with the non-specific ATR inhibitor caffeine or depletion of VPRBP by siRNA did not inhibit formation of Vpr nuclear foci, mutations in the C-terminal domain of Vpr and cytoplasmic sequestration of Vpr by overexpression of Gag-Pol resulted in impaired formation of these nuclear structures and defective G2 arrest. Consistently, we observed that G2 arrest-competent sooty mangabey Vpr could form these foci but not its G2 arrest-defective paralog Vpx, suggesting that formation of Vpr nuclear foci represents a critical early event in the induction of G2 arrest. Indeed, we found that Vpr could associate to chromatin via its C-terminal domain and that it could form a complex with VPRBP on chromatin. Finally, analysis of Vpr nuclear foci by time-lapse microscopy showed that they were highly mobile and stable structures. Overall, our results suggest that Vpr recruits the DDB1-CUL4A (VPRBP) E3 ligase to these nuclear foci and uses these mobile structures to target a chromatin-bound cellular substrate for ubiquitination in order to induce DNA damage/replication stress, ultimately leading to ATR activation and G2 cell cycle arrest

    Reducing the environmental impact of surgery on a global scale: systematic review and co-prioritization with healthcare workers in 132 countries

    Get PDF
    Abstract Background Healthcare cannot achieve net-zero carbon without addressing operating theatres. The aim of this study was to prioritize feasible interventions to reduce the environmental impact of operating theatres. Methods This study adopted a four-phase Delphi consensus co-prioritization methodology. In phase 1, a systematic review of published interventions and global consultation of perioperative healthcare professionals were used to longlist interventions. In phase 2, iterative thematic analysis consolidated comparable interventions into a shortlist. In phase 3, the shortlist was co-prioritized based on patient and clinician views on acceptability, feasibility, and safety. In phase 4, ranked lists of interventions were presented by their relevance to high-income countries and low–middle-income countries. Results In phase 1, 43 interventions were identified, which had low uptake in practice according to 3042 professionals globally. In phase 2, a shortlist of 15 intervention domains was generated. In phase 3, interventions were deemed acceptable for more than 90 per cent of patients except for reducing general anaesthesia (84 per cent) and re-sterilization of ‘single-use’ consumables (86 per cent). In phase 4, the top three shortlisted interventions for high-income countries were: introducing recycling; reducing use of anaesthetic gases; and appropriate clinical waste processing. In phase 4, the top three shortlisted interventions for low–middle-income countries were: introducing reusable surgical devices; reducing use of consumables; and reducing the use of general anaesthesia. Conclusion This is a step toward environmentally sustainable operating environments with actionable interventions applicable to both high– and low–middle–income countries

    HIV-1 Vpr Induces the K48-Linked Polyubiquitination and Proteasomal Degradation of Target Cellular Proteins To Activate ATR and Promote G2 Arrest ▿ †

    No full text
    HIV-1 viral protein R (Vpr) induces cell cycle arrest at the G2/M phase by a mechanism involving the activation of the DNA damage sensor ATR. We and others recently showed that Vpr performs this function by subverting the activity of the DDB1-CUL4A (VPRBP) E3 ubiquitin ligase. Vpr could thus act as a connector between the E3 ligase and an unknown cellular factor whose ubiquitination would induce G2 arrest. While attractive, this model is based solely on the indirect observation that some mutants of Vpr retain their interaction with the E3 ligase but fail to induce G2 arrest. Using a tandem affinity purification approach, we observed that Vpr interacts with ubiquitinated cellular proteins and that this association requires the recruitment of an active E3 ligase given that the depletion of VPRBP by RNA interference or the overexpression of a dominant negative mutant of CUL4A decreased this association. Importantly, G2-arrest-defective mutants of Vpr in the C-terminal putative substrate-interacting domain displayed a decreased association with ubiquitinated proteins. We also found that the inhibition of proteasomal activity increased this association and that the ubiquitin chains were at least in part constituted of classical K48 linkages. Interestingly, the inhibition of K48 polyubiquitination specifically impaired the Vpr-induced phosphorylation of H2AX, an early target of ATR, but did not affect UV-induced H2AX phosphorylation. Overall, our results provide direct evidence that the association of Vpr with the DDB1-CUL4A (VPRBP) E3 ubiquitin ligase induces the K48-linked polyubiquitination of as-yet-unknown cellular proteins, resulting in their proteasomal degradation and ultimately leading to the activation of ATR and G2 arrest

    Defining the Interactions and Role of DCAF1/VPRBP in the DDB1-Cullin4A E3 Ubiquitin Ligase Complex Engaged by HIV-1 Vpr to Induce a G<sub>2</sub> Cell Cycle Arrest

    No full text
    <div><p>HIV viral protein R (Vpr) induces a cell cycle arrest at the G<sub>2</sub>/M phase by activating the ATR DNA damage/replication stress signalling pathway through engagement of the DDB1-CUL4A-DCAF1 E3 ubiquitin ligase via a direct binding to the substrate specificity receptor DCAF1. Since no high resolution structures of the DDB1-DCAF1-Vpr substrate recognition module currently exist, we used a mutagenesis approach to better define motifs in DCAF1 that are crucial for Vpr and DDB1 binding. Herein, we show that the minimal domain of DCAF1 that retained the ability to bind Vpr and DDB1 was mapped to residues 1041 to 1393 (DCAF1 WD). Mutagenic analyses identified an α-helical H-box motif and F/YxxF/Y motifs located in the N-terminal domain of DCAF1 WD that are involved in exclusive binding to DDB1. While we could not identify elements specifically involved in Vpr binding, overall, the mutagenesis data suggest that the predicted ÎČ-propeller conformation of DCAF1 is likely to be critical for Vpr association. Importantly, we provide evidence that binding of Vpr to DCAF1 appears to modulate the formation of a DDB1/DCAF1 complex. Lastly, we show that expression of DCAF1 WD in the absence of endogenous DCAF1 was not sufficient to enable Vpr-mediated G<sub>2</sub> arrest activity. Overall, our results reveal that Vpr and DDB1 binding on DCAF1 can be genetically separated and further suggest that DCAF1 contains determinants in addition to the Vpr and DDB1 minimal binding domain, which are required for Vpr to enable the induction of a G<sub>2</sub> arrest.</p></div

    Delineation of the minimal domain of DCAF1 that interacts with HIV-1 Vpr and DDB1.

    No full text
    <p><b>A.</b> Schematic representation of Myc-DCAF1 WT (1-1507), Myc-DCAF1 WD (1041-1393) and Myc-DCAF1 1377 (1041-1377). The different domains of DCAF1 with their amino-acid positions are highlighted. Additionally, the region targeted by the full length DCAF1-specific bp3 siRNA is highlighted in red (see below). <b>B-C</b>. HEK293T cells were mock-transfected (lanes 1 and 2) or transfected with Myc-DCAF1 (1-1507) (lanes 3 and 4), Myc-DCAF1 WD (1041-1393) (lanes 5 and 6) or with Myc-DCAF1 1377 (1041-1377) (lanes 7 and 8) -encoding plasmids in the presence of empty vector (lanes 1, 3, 5 and 7) or HA-tagged Vpr-expressing plasmid (lanes 2, 4, 6, and 8). Total amounts of DNA were adjusted with empty vector so that similar quantities of plasmids were transfected in each sample. <b>B.</b> Immunoprecipitations using anti-Myc antibody were performed on cell extracts using protein-A sepharose beads. The levels of HA-Vpr, endogenous DDB1, Myc-DCAF1 proteins and actin were monitored in cell extracts as well as, when applicable, in immunoprecipitated fractions by Western Blot using specific antibodies. <b>C.</b> Quantitation of DDB1 binding efficiency. Band signals corresponding to DDB1 in immunocomplexes were scanned by laser densitometry. The ratio of DDB1 signal over that of precipitated Myc-DCAF1 1507 or Myc-DCAF1WD was calculated and expressed as the percentage of that obtained in the absence of Vpr, which was assigned a value of 100%. Error bars indicate the standard error of the mean (SEM) from the quantitative analysis of three independent experiments. Statistical analysis was performed as described in the Experimental Procedures (p<0.05; ns; non significant). <b>D</b>. Immunoprecipitation using anti-HA antibody was performed on cell extracts using anti-HA antibody-coupled agarose beads. The levels of HA-Vpr, endogenous DDB1, endogenous DCAF1, Myc-DCAF1 proteins and actin were monitored in cell extracts as well as, when applicable, in immunoprecipitated fractions by Western Blot using specific antibodies. The data shown here are representative of results obtained in three independent experiments. * denotes the light chain of the IgG used for immunoprecipitation. # represents non-specific immunoprecipitated proteins. <b>E.</b> Structural and molecular features of the DCAF1 WD minimal domain. Consensus secondary structure prediction of DCAF1 WD 1041-1393 was generated using the PSI-PRED server and structural data obtained from the 3D modelization. Orange lines highlight the predicted ÎČ-sheet structures while the green line and the green amino-acid residues highlight α-helices and the putative H-box motif (see below), respectively. The F/YxxF/Y repeats are highlighted in purple whereas the WDxR motifs are highlighted in red.</p

    DCAF1 does not solely act as a bridge to engage Vpr to the DDB1-CRL4A E3 ubiquitin ligase and induce G<sub>2</sub> cell cycle arrest.

    No full text
    <p>A-B. HEK293T cells were mock-transfected (lanes 2 and 3) or transfected with HA-Vpr-expressing plasmid (lanes 4 and 5) or transfected with DCAF1 bp3R (DCAF1 1-1507 bp3 siRNA resistant) (lanes 6 and 7), Myc-DCAF1 WD (lanes 8 and 9) or Myc-DCAF1 1377 (lanes 10-11)-encoding plasmids in the presence of HA-Vpr-expressing plasmid. All cells were also transfected with a plasmid encoding GFP and treated with either non-targeting siRNA (<i>NT siRNA</i>) (lanes 2, 4, 6, 8 and 10) or specific DCAF1 bp3 siRNA (<i>bp3 siRNA</i>) (lanes 3, 5, 7, 9 and 11). Non-transfected HEK293T cells were used as negative control (lane 1). <b>A.</b> Non-transfected or transfected HEK293T cells were lysed in 0.5% Triton lysis buffer and subjected to anti-HA or anti-Myc immunoprecipitation and further resolved on SDS-PAGE. The level of HA-Vpr, actin, exogenous and endogenous DCAF1, endogenous DDB1, Myc-DCAF1 WD and Myc-DCAF1 1377 were monitored in cell extracts as well as in the immunoprecipitated fractions by Western Blot using specific antibodies. * denotes the light chain of the IgG used for immunoprecipitation. # represents non-specific immunoprecipitated proteins. <b>B.</b> Cell cycle profile of transfected cells (GFP+) analyzed in A. G<sub>2</sub>/M:G<sub>1</sub> ratios were determined using the Modfit Software. <b>C.</b> The graph depicts the mean G<sub>2</sub>/M:G<sub>1</sub> ratios obtained in two independent experiments. Errors bars represent SEM.</p
    corecore