31 research outputs found

    CRISPR/Cas9-constructed pseudorabies virus mutants reveal the importance of UL13 in alphaherpesvirus escape from genome silencing

    Get PDF
    Latent and recurrent productive infection of long-living cells, such as neurons, enables alphaherpesviruses to persist in their host populations. Still, the viral factors involved in these events remain largely obscure. Using a complementation assay in compartmented primary peripheral nervous system (PNS) neuronal cultures, we previously reported that productive replication of axonally delivered genomes is facilitated by pseudorabies virus (PRV) tegument proteins. Here, we sought to unravel the role of tegument protein UL13 in this escape from silencing. We first constructed four new PRV mutants in the virulent Becker strain using CRISPR/Cas9-mediated gene replacement: (i) PRV Becker defective for UL13 expression (PRV Delta UL13), (ii) PRV where UL13 is fused to eGFP (PRV UL13-eGFP), and two control viruses (iii and iv) PRV where VP16 is fused with mTurquoise at either the N terminus (PRV mTurq-VP16) or the C terminus (PRV VP16-mTurq). Live-cell imaging of PRV capsids showed efficient retrograde transport after axonal infection with PRV UL13-eGFP, although we did not detect dual-color particles. However, immunofluorescence staining of particles in mid-axons indicated that UL13 might be cotransported with PRV capsids in PNS axons. Superinfecting nerve cell bodies with UV-inactivated PRV DUL13 failed to efficiently promote escape from genome silencing compared to UV-PRV wild type and UV-PRV UL13-eGFP superinfection. However, UL13 does not act directly in the escape from genome silencing, as adeno-associated virus (AAV)-mediated UL13 expression in neuronal cell bodies was not sufficient to provoke escape from genome silencing. Based on this, we suggest that UL13 may contribute to initiation of productive infection through phosphorylation of other tegument proteins. IMPORTANCE Alphaherpesviruses have mastered various strategies to persist in an immunocompetent host, including the induction of latency and reactivation in peripheral nervous system (PNS) ganglia. We recently discovered that the molecular mechanism underlying escape from latency by the alphaherpesvirus pseudorabies virus (PRV) relies on a structural viral tegument protein. This study aimed at unravelling the role of tegument protein UL13 in PRV escape from latency. First, we confirmed the use of CRISPR/Cas9-mediated gene replacement as a versatile tool to modify the PRV genome. Next, we used our new set of viral mutants and AAV vectors to conclude the indirect role of UL13 in PRV escape from latency in primary neurons, along with its spatial localization during retrograde capsid transport in axons. Based on these findings, we speculate that UL13 phosphorylates one or more tegument proteins, thereby priming these putative proteins to induce escape from genome silencing

    A Pseudorabies Virus Serine/Threonine Kinase, US3, Promotes Retrograde Transport in Axons via Akt/mToRC1

    No full text
    Infection of peripheral axons by alpha herpesviruses (AHVs) is a critical stage in establishing a lifelong infection in the host. Upon entering the cytoplasm of axons, AHV nucleocapsids and associated inner-tegument proteins must engage the cellular retrograde transport machinery to promote the long-distance movement of virion components to the nucleus. The current model outlining this process is incomplete, and further investigation is required to discover all viral and cellular determinants involved as well as the temporality of the events. Using a modified trichamber system, we have discovered a novel role of the pseudorabies virus (PRV) serine/threonine kinase US3 in promoting efficient retrograde transport of nucleocapsids. We discovered that transporting nucleocapsids move at similar velocities in both the presence and absence of a functional US3 kinase; however, fewer nucleocapsids are moving when US3 is absent, and they move for shorter periods of time before stopping, suggesting that US3 is required for efficient nucleocapsid engagement with the retrograde transport machinery. This led to fewer nucleocapsids reaching the cell bodies to produce a productive infection 12 h later. Furthermore, US3 was responsible for the induction of local translation in axons as early as 1 h postinfection (hpi) through the stimulation of a phosphatidylinositol 3-kinase (PI3K)/Akt-mToRC1 pathway. These data describe a novel role for US3 in the induction of local translation in axons during AHV infection, a critical step in transport of nucleocapsids to the cell body. IMPORTANCE Neurons are highly polarized cells with axons that can reach centimeters in length. Communication between axons at the periphery and the distant cell body is a relatively slow process involving the active transport of chemical messengers. There is a need for axons to respond rapidly to extracellular stimuli. Translation of repressed mRNAs present within the axon occurs to enable rapid, localized responses independently of the cell body. AHVs have evolved a way to hijack local translation in the axons to promote their transport to the nucleus. We have determined the cellular mechanism and viral components involved in the induction of axonal translation. The US3 serine/threonine kinase of PRV activates Akt-mToRC1 signaling pathways early during infection to promote axonal translation. When US3 is not present, the number of moving nucleocapsids and their processivity are reduced, suggesting that US3 activity is required for efficient engagement of nucleocapsids with the retrograde transport machinery

    Interferon-λ Activates a Differential Response in Peripheral Neurons That Is Effective against Alpha Herpesvirus Infections

    No full text
    Alpha herpesviruses (α-HV) infect host mucosal epithelial cells prior to establishing a life-long latent infection in the peripheral nervous system. The initial spread of viral particles from mucosa to the nervous system and the role of intrinsic immune responses at this barrier is not well understood. Using primary neurons cultured in compartmentalized chambers, prior studies performed on Pseudorabies virus (PRV) have demonstrated that type I and type II interferons (IFNs) induce a local antiviral response in axons via distinct mechanisms leading to a reduction in viral particle transport to the neuronal nucleus. A new class of interferons known as type III IFNs has been shown to play an immediate role against viral infection in mucosal epithelial cells. However, the antiviral effects of type III IFNs within neurons during α-HV infection are largely unknown. In this study, we focused on elucidating the antiviral activity of type III IFN against PRV neuronal infection, and we compared the interferon-stimulated gene (ISGs) induction pattern in neurons to non-neuronal cells. We found that IFN pre-exposure of both primary neurons and fibroblast cells significantly reduces PRV virus yield, albeit by differential STAT activation and ISG induction patterns. Notably, we observed that type III IFNs trigger the expression of a subset of ISGs mainly through STAT1 activation to induce an antiviral state in primary peripheral neurons

    Low MOI axonal PRV infection in compartmented neuronal cultures results in a quiescent infection in a small number of neurons.

    No full text
    <p><b>(A)</b> Tri-chamber compartments, S: soma (cell bodies), M: methocel (middle), N: axonal. PRV 233 infection was made in the N compartment at an MOI of 0.01. DiI (red) was added to the N compartment media 6 hpi to label red cell bodies that project axons to the N compartment. Circles highlight primarily infected neurons. Insets show single infected neurons (green) among surrounding non-infected neurons. <b>(B)</b> GFP positive, DiI labeled or non-labeled cell bodies were counted at 3 dpi (separate or merged channels are shown, arrowhead points to one GFP positive cell body, ph: phase contrast). Ratios of either red cell bodies to all S compartment neurons (connectivity), or green cell bodies to dual color (green and red) cell bodies (infectivity) were calculated and shown in the graph. <b>(C)</b> RNA was isolated after 7 days of either S compartment or N compartment infection with PRV180 at an MOI of 0.01. LAT and EP0 transcripts were quantitated. S compartment (cell body) infection resulted in a productive infection (red capsid accumulation in all of the cells), whereas N compartment (axonal) infection was silent (no detectable red fluorescent signal anywhere in the S compartment). The graph shows the ratio of each transcript in axonal to cell body infection after normalization to 28S rRNA. <b>(D)</b> Illustration of the complementation assay: N compartments are infected with PRV180 at an MOI of 0.01 while S compartments are treated with drugs, inhibitors, viruses or virus-like particles.</p

    Simultaneous infection of cell bodies with UVPRV enables axonally infecting PRV180 to escape from silencing.

    No full text
    <p><b>(A)</b> Control and UVPRV959 (MOI 10) complemented S compartments are shown (3 dpi). Red channel is shown after background filtration. Raw image is used for the green channel (no signal). <b>(B)</b> Quantitation of imaging data was graphed including control dishes (7 dpi), UV959 (3 dpi), UVgBnull or UVgDnull PRV complementation (7 dpi), and UV959 plus H89 (UVH89) complementation (3 dpi). Each data point represents one dish (ns is not significant and **** is p<0.0001). <b>(C)</b> Western blot analysis of PKA targets after 3 hours of PRV180 and UVPRV infection in S compartments at an MOI of 10 with (+) or without (-) H89 treatment are shown in comparison to forskolin and dbcAMP treatments. <b>(D)</b> Control dishes with axonal PRV180 infection (MOI 0.01) were harvested either 3 or 10 dpi. S compartments infected only with UVPRV (MOI 10), and complementation assay samples were harvested at 3 dpi. The presence of viral capsid (VP5) and tegument (UL47 and EP0) proteins was determined by WB using half of the lysates (each lane represents contents of one S compartment). Beta-actin was used as a loading control. The other half of the lysates was used to quantitate the amount of viral DNA. <b>(E)</b> PRV DNA was quantitated using UL54 primers. DNA amounts calculated based on threshold cycle values are shown in the graph. Red line shows the detection limit.</p
    corecore