23 research outputs found

    The pUL37 tegument protein guides alphaherpesvirus retrograde axonal transport to promote neuroinvasion

    Get PDF
    A hallmark property of the neurotropic alpha-herpesvirinae is the dissemination of infection to sensory and autonomic ganglia of the peripheral nervous system following an initial exposure at mucosal surfaces. The peripheral ganglia serve as the latent virus reservoir and the source of recurrent infections such as cold sores (herpes simplex virus type I) and shingles (varicella zoster virus). However, the means by which these viruses routinely invade the nervous system is not fully understood. We report that an internal virion component, the pUL37 tegument protein, has a surface region that is an essential neuroinvasion effector. Mutation of this region rendered herpes simplex virus type 1 (HSV-1) and pseudorabies virus (PRV) incapable of spreading by retrograde axonal transport to peripheral ganglia both in culture and animals. By monitoring the axonal transport of individual viral particles by time-lapse fluorescence microscopy, the mutant viruses were determined to lack the characteristic sustained intracellular capsid motion along microtubules that normally traffics capsids to the neural soma. Consistent with the axonal transport deficit, the mutant viruses did not reach sites of latency in peripheral ganglia, and were avirulent. Despite this, viral propagation in peripheral tissues and in cultured epithelial cell lines remained robust. Selective elimination of retrograde delivery to the nervous system has long been sought after as a means to develop vaccines against these ubiquitous, and sometimes devastating viruses. In support of this potential, we find that HSV-1 and PRV mutated in the effector region of pUL37 evoked effective vaccination against subsequent nervous system challenges and encephalitic disease. These findings demonstrate that retrograde axonal transport of the herpesviruses occurs by a virus-directed mechanism that operates by coordinating opposing microtubule motors to favor sustained retrograde delivery of the virus to the peripheral ganglia. The ability to selectively eliminate the retrograde axonal transport mechanism from these viruses will be useful in trans-synaptic mapping studies of the mammalian nervous system, and affords a new vaccination paradigm for human and veterinary neurotropic herpesviruses

    Simplified Models for LHC New Physics Searches

    Get PDF
    This document proposes a collection of simplified models relevant to the design of new-physics searches at the LHC and the characterization of their results. Both ATLAS and CMS have already presented some results in terms of simplified models, and we encourage them to continue and expand this effort, which supplements both signature-based results and benchmark model interpretations. A simplified model is defined by an effective Lagrangian describing the interactions of a small number of new particles. Simplified models can equally well be described by a small number of masses and cross-sections. These parameters are directly related to collider physics observables, making simplified models a particularly effective framework for evaluating searches and a useful starting point for characterizing positive signals of new physics. This document serves as an official summary of the results from the "Topologies for Early LHC Searches" workshop, held at SLAC in September of 2010, the purpose of which was to develop a set of representative models that can be used to cover all relevant phase space in experimental searches. Particular emphasis is placed on searches relevant for the first ~50-500 pb-1 of data and those motivated by supersymmetric models. This note largely summarizes material posted at http://lhcnewphysics.org/, which includes simplified model definitions, Monte Carlo material, and supporting contacts within the theory community. We also comment on future developments that may be useful as more data is gathered and analyzed by the experiments.Comment: 40 pages, 2 figures. This document is the official summary of results from "Topologies for Early LHC Searches" workshop (SLAC, September 2010). Supplementary material can be found at http://lhcnewphysics.or

    Remdesivir Inhibits SARS-CoV-2 in Human Lung Cells and Chimeric SARS-CoV Expressing the SARS-CoV-2 RNA Polymerase in Mice

    Get PDF
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the novel viral disease COVID-19. With no approved therapies, this pandemic illustrates the urgent need for broad-spectrum antiviral countermeasures against SARS-CoV-2 and future emerging CoVs. We report that remdesivir (RDV) potently inhibits SARS-CoV-2 replication in human lung cells and primary human airway epithelial cultures (EC50 = 0.01 ÎĽM). Weaker activity is observed in Vero E6 cells (EC50 = 1.65 ÎĽM) because of their low capacity to metabolize RDV. To rapidly evaluate in vivo efficacy, we engineered a chimeric SARS-CoV encoding the viral target of RDV, the RNA-dependent RNA polymerase of SARS-CoV-2. In mice infected with the chimeric virus, therapeutic RDV administration diminishes lung viral load and improves pulmonary function compared with vehicle-treated animals. These data demonstrate that RDV is potently active against SARS-CoV-2 in vitro and in vivo, supporting its further clinical testing for treatment of COVID-19

    The pUL37 tegument protein guides alphaherpesvirus retrograde axonal transport to promote neuroinvasion

    Get PDF
    A hallmark property of the neurotropic alpha-herpesvirinae is the dissemination of infection to sensory and autonomic ganglia of the peripheral nervous system following an initial exposure at mucosal surfaces. The peripheral ganglia serve as the latent virus reservoir and the source of recurrent infections such as cold sores (herpes simplex virus type I) and shingles (varicella zoster virus). However, the means by which these viruses routinely invade the nervous system is not fully understood. We report that an internal virion component, the pUL37 tegument protein, has a surface region that is an essential neuroinvasion effector. Mutation of this region rendered herpes simplex virus type 1 (HSV-1) and pseudorabies virus (PRV) incapable of spreading by retrograde axonal transport to peripheral ganglia both in culture and animals. By monitoring the axonal transport of individual viral particles by time-lapse fluorescence microscopy, the mutant viruses were determined to lack the characteristic sustained intracellular capsid motion along microtubules that normally traffics capsids to the neural soma. Consistent with the axonal transport deficit, the mutant viruses did not reach sites of latency in peripheral ganglia, and were avirulent. Despite this, viral propagation in peripheral tissues and in cultured epithelial cell lines remained robust. Selective elimination of retrograde delivery to the nervous system has long been sought after as a means to develop vaccines against these ubiquitous, and sometimes devastating viruses. In support of this potential, we find that HSV-1 and PRV mutated in the effector region of pUL37 evoked effective vaccination against subsequent nervous system challenges and encephalitic disease. These findings demonstrate that retrograde axonal transport of the herpesviruses occurs by a virus-directed mechanism that operates by coordinating opposing microtubule motors to favor sustained retrograde delivery of the virus to the peripheral ganglia. The ability to selectively eliminate the retrograde axonal transport mechanism from these viruses will be useful in trans-synaptic mapping studies of the mammalian nervous system, and affords a new vaccination paradigm for human and veterinary neurotropic herpesviruses

    The pUL37 tegument protein guides alpha-herpesvirus retrograde axonal transport to promote neuroinvasion.

    No full text
    A hallmark property of the neurotropic alpha-herpesvirinae is the dissemination of infection to sensory and autonomic ganglia of the peripheral nervous system following an initial exposure at mucosal surfaces. The peripheral ganglia serve as the latent virus reservoir and the source of recurrent infections such as cold sores (herpes simplex virus type I) and shingles (varicella zoster virus). However, the means by which these viruses routinely invade the nervous system is not fully understood. We report that an internal virion component, the pUL37 tegument protein, has a surface region that is an essential neuroinvasion effector. Mutation of this region rendered herpes simplex virus type 1 (HSV-1) and pseudorabies virus (PRV) incapable of spreading by retrograde axonal transport to peripheral ganglia both in culture and animals. By monitoring the axonal transport of individual viral particles by time-lapse fluorescence microscopy, the mutant viruses were determined to lack the characteristic sustained intracellular capsid motion along microtubules that normally traffics capsids to the neural soma. Consistent with the axonal transport deficit, the mutant viruses did not reach sites of latency in peripheral ganglia, and were avirulent. Despite this, viral propagation in peripheral tissues and in cultured epithelial cell lines remained robust. Selective elimination of retrograde delivery to the nervous system has long been sought after as a means to develop vaccines against these ubiquitous, and sometimes devastating viruses. In support of this potential, we find that HSV-1 and PRV mutated in the effector region of pUL37 evoked effective vaccination against subsequent nervous system challenges and encephalitic disease. These findings demonstrate that retrograde axonal transport of the herpesviruses occurs by a virus-directed mechanism that operates by coordinating opposing microtubule motors to favor sustained retrograde delivery of the virus to the peripheral ganglia. The ability to selectively eliminate the retrograde axonal transport mechanism from these viruses will be useful in trans-synaptic mapping studies of the mammalian nervous system, and affords a new vaccination paradigm for human and veterinary neurotropic herpesviruses

    Mice immunized with the HSV-1 R2 mutant are protected from HSV-1 neuroinvasion and periocular skin disease.

    No full text
    <p><b>(A)</b> Mice were vaccinated by inoculating the left cornea with 7 μl of the HSV-1 R2 mutant stock (R2; 9 x 10<sup>7</sup> PFU/ml). Sham vaccinated animals received an equivalent volume of conditioned media. At 14 days post vaccination mice were challenged with 7 μl of wild-type HSV-1 strain F (1.0 x 10<sup>8</sup> PFU/ml) in the right eye. Mice were euthanized at 4 days post infection and the viral load in the whole ipsilateral trigeminal ganglia and brain were determined. The mean titer of each data set is indicated by a red bar (n.d., not detected). <b>(B)</b> Mice were vaccinated as described in (A), at 14 days post vaccination mice were challenged with 5 μl of wild-type HSV-1 McKrae (6.0 x 10<sup>8</sup> PFU/ml) in the right eye and monitored for survival. <b>(C)</b> Mice were vaccinated as described in (A), and at 14 days post vaccination mice were challenged in the right eye with wild-type HSV-1 (either strain F or McKrae as indicated). The right eye of vaccinated animals was scored for periocular skin disease at the indicated day post challenge. Scoring was based on previous published criteria [<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006741#ppat.1006741.ref056" target="_blank">56</a>]: 0, no lesions; 1, minimal eyelid swelling; 2, moderate eye lid swelling; 3, severe eye lid swelling with no periocular hair loss; 4, eyes swollen shut with minimal ocular discharge and periocular hair loss; 5 eyes swollen shut with severe periocular hair loss and skin lesions. Values are mean disease scores ± s.d. * Mice were euthanized at 5 days post challenge due to pronounced neurological symptoms.</p

    The PRV pUL37 R2 region is essential for virulence and can be mutated without causing misfolding of the surrounding protein structure.

    No full text
    <p><b>(A)</b> Kaplan–Meier presentation of mouse survival following intranasal instillation of wild-type PRV (WT) or PRV carrying mutations in the R1, R2, or R3 regions of the pUL37 tegument protein (n = 5 animals for each virus). All viruses encode a mCherry tag fused to the pUL25 capsid protein as previously described [<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006741#ppat.1006741.ref030" target="_blank">30</a>,<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006741#ppat.1006741.ref047" target="_blank">47</a>]. <b>(B)</b> The crystal structure of the N-terminal half of the PRV pUL37 R2 mutant (R2; lilac), determined in this work, was overlaid onto the previously determined wild-type structure (WT; beige) [<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006741#ppat.1006741.ref047" target="_blank">47</a>] with rmsd 0.5538 Å over 479 aligned residues as determined in Coot [<a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1006741#ppat.1006741.ref093" target="_blank">93</a>]. A close-up view of R2 is shown to the right with the side chains of the five targeted amino acids indicated for wild type and the mutant.</p

    pUL37 R2 is essential for HSV-1 neuroinvasion.

    No full text
    <p><b>(A)</b> Diagram of the neuroinvasive route examined in mice following inoculation of a scarified cornea with HSV-1. <b>(B)</b> Representative images of the cornea (2 dpi), trigeminal ganglia (6 dpi), and principal sensory trigeminal nuclei (Pr5; 6 dpi), following corneal inoculation with wild-type or R2-mutant HSV-1. Infected cells were visualized by virtue of a pUL25/mCherry fluorescent capsid reporter encoded by the viruses (scale bars for trigeminal ganglia and Pr5 are 500 ÎĽm; cornea scale bar is 1000 ÎĽm). <b>(C)</b> Mice were infected on both eyes with untagged wild-type (WT) or R2-mutant (R2) HSV-1 following corneal scarification. Viral titers in the tear films were independently determined from each eye by swabbing at the indicated day post-infection (dpi). At 4 days post corneal inoculation, the combined titer of the left and right trigeminal ganglia and of the whole brain were determined. The mean titer of each data set is indicated by a red bar (5 mice per virus; *, p < 0.05 based on a two-tailed unpaired <i>t</i> test). <b>(D)</b> qPCR was preformed using primers directed against the HSV-1 UL35 gene. Trigeminal ganglia and brain samples were collected at 4 days post infection and scored as positive for viral DNA if the threshold cycle (Ct) value was below the average Ct value of the water controls by more than two standard deviations (inset). Amplification curves for both HSV-1 WT and R2-mutant infected samples are shown.</p
    corecore