Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Cell Sci ; 136(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36093836

ABSTRACT

Intracellular mature viruses (IMVs) are the first and most abundant infectious form of vaccinia virus to assemble during its replication cycle. IMVs can undergo microtubule-based motility, but their directionality and the motor involved in their transport remain unknown. Here, we demonstrate that IMVs, like intracellular enveloped viruses (IEVs), the second form of vaccinia that are wrapped in Golgi-derived membranes, recruit kinesin-1 and undergo anterograde transport. In vitro reconstitution of virion transport in infected cell extracts revealed that IMVs and IEVs move toward microtubule plus ends with respective velocities of 0.66 and 0.56 µm/s. Quantitative imaging established that IMVs and IEVs recruit an average of 139 and 320 kinesin-1 motor complexes, respectively. In the absence of kinesin-1, there was a near-complete loss of in vitro motility and reduction in the intracellular spread of both types of virions. Our observations demonstrate that kinesin-1 transports two morphologically distinct forms of vaccinia. Reconstitution of vaccinia-based microtubule motility in vitro provides a new model to elucidate how motor number and regulation impacts transport of a bona fide kinesin-1 cargo.


Subject(s)
Kinesins , Vaccinia , Cell Extracts , Humans , Microtubules/metabolism , Vaccinia/metabolism , Vaccinia virus , Virion/physiology
2.
Proc Natl Acad Sci U S A ; 117(27): 15935-15946, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32571912

ABSTRACT

Excessive tumor necrosis factor (TNF) is known to cause significant pathology. Paradoxically, deficiency in TNF (TNF-/-) also caused substantial pathology during respiratory ectromelia virus (ECTV) infection, a surrogate model for smallpox. TNF-/- mice succumbed to fulminant disease whereas wild-type mice, and those engineered to express only transmembrane TNF (mTNF), fully recovered. TNF deficiency did not affect viral load or leukocyte recruitment but caused severe lung pathology and excessive production of the cytokines interleukin (IL)-6, IL-10, transforming growth factor beta (TGF-ß), and interferon gamma (IFN-γ). Short-term blockade of these cytokines significantly reduced lung pathology in TNF-/- mice concomitant with induction of protein inhibitor of activated STAT3 (PIAS3) and/or suppressor of cytokine signaling 3 (SOCS3), factors that inhibit STAT3 activation. Consequently, inhibition of STAT3 activation with an inhibitor reduced lung pathology. Long-term neutralization of IL-6 or TGF-ß protected TNF-/- mice from an otherwise lethal infection. Thus, mTNF alone is necessary and sufficient to regulate lung inflammation but it has no direct antiviral activity against ECTV. The data indicate that targeting specific cytokines or cytokine-signaling pathways to reduce or ameliorate lung inflammation during respiratory viral infections is possible but that the timing and duration of the interventive measure are critical.


Subject(s)
Cytokines/metabolism , Poxviridae Infections/virology , Poxviridae/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line, Tumor , Female , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Poxviridae/immunology , Poxviridae Infections/immunology , Poxviridae Infections/pathology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Transforming Growth Factor beta/metabolism
3.
Cell Microbiol ; 22(8): e13206, 2020 08.
Article in English | MEDLINE | ID: mdl-32237038

ABSTRACT

The induction of Smad signalling by the extracellular ligand TGF-ß promotes tissue plasticity and cell migration in developmental and pathological contexts. Here, we show that vaccinia virus (VACV) stimulates the activity of Smad transcription factors and expression of TGF-ß/Smad-responsive genes at the transcript and protein levels. Accordingly, infected cells share characteristics to those undergoing TGF-ß/Smad-mediated epithelial-to-mesenchymal transition (EMT). Depletion of the Smad4 protein, a common mediator of TGF-ß signalling, results in an attenuation of viral cell-to-cell spread and reduced motility of infected cells. VACV induction of TGF-ß/Smad-responsive gene expression does not require the TGF-ß ligand or type I and type II TGF-ß receptors, suggesting a novel, non-canonical Smad signalling pathway. Additionally, the spread of ectromelia virus, a related orthopoxvirus that does not activate a TGF-ß/Smad response, is enhanced by the addition of exogenous TGF-ß. Together, our results indicate that VACV orchestrates a TGF-ß-like response via a unique activation mechanism to enhance cell migration and promote virus spread.


Subject(s)
Signal Transduction , Smad4 Protein/genetics , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Vaccinia virus/physiology , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition , HT29 Cells , HaCaT Cells , HeLa Cells , Humans , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , Vaccinia virus/drug effects
4.
J Virol ; 93(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30626686

ABSTRACT

Live viral vaccines elicit protective, long-lived humoral immunity, but the underlying mechanisms through which this occurs are not fully elucidated. Generation of affinity matured, long-lived protective antibody responses involve close interactions between T follicular helper (TFH) cells, germinal center (GC) B cells, and T follicular regulatory (TFR) cells. We postulated that escalating concentrations of antigens from replicating viruses or live vaccines, spread through the hematogenous route, are essential for the induction and maintenance of long-lived protective antibody responses. Using replicating and poorly replicating or nonreplicating orthopox and influenza A viruses, we show that the magnitude of TFH cell, GC B cell, and neutralizing antibody responses is directly related to virus replicative capacity. Further, we have identified that both lymphoid and circulating TFH:TFR cell ratios during the peak GC response can be used as an early predictor of protective, long-lived antibody response induction. Finally, administration of poorly or nonreplicating viruses to allow hematogenous spread generates significantly stronger TFH:TFR ratios and robust TFH, GC B cell and neutralizing antibody responses.IMPORTANCE Neutralizing antibody response is the best-known correlate of long-term protective immunity for most of the currently licensed clinically effective viral vaccines. However, the host immune and viral factors that are critical for the induction of robust and durable antiviral humoral immune responses are not well understood. Our study provides insight into the dynamics of key cellular mediators of germinal center reaction during live virus infections and the influence of viral replicative capacity on the magnitude of antiviral antibody response and effector function. The significance of our study lies in two key findings. First, the systemic spread of even poorly replicating or nonreplicating viruses to mimic the spread of antigens from replicating viruses due to escalating antigen concentration is fundamental to the induction of durable antibody responses. Second, the TFH:TFR ratio may be used as an early predictor of protective antiviral humoral immune responses long before memory responses are generated.


Subject(s)
Antibodies, Neutralizing/immunology , Antigens/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Virus Replication/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation/immunology , B-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Line , Chlorocebus aethiops , Dogs , Germinal Center/immunology , Immunity, Humoral/immunology , Madin Darby Canine Kidney Cells , Mesocricetus , Mice , Mice, Inbred C57BL , Vaccines, Attenuated/immunology
5.
Semin Cell Dev Biol ; 46: 155-63, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26459972

ABSTRACT

Actin nucleation drives a diversity of critical cellular processes and the motility of a select group of viral pathogens. Vaccinia virus and baculovirus, Autographa californica multiple nucleopolyhedrovirus, recruit and activate the cellular actin nucleator, the Arp2/3 complex, at the surface of virus particles thereby instigating highly localized actin nucleation. The extension of these filaments provides a mechanical force that bestows the ability to navigate the intracellular environment and promote their infectious cycles. This review outlines the viral and cellular proteins that initiate and regulate the signalling networks leading to viral modification of the actin cytoskeleton and summarizes recent insights into the role of actin-based virus transport.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Nucleopolyhedroviruses/metabolism , Vaccinia virus/metabolism , Virus Diseases/metabolism , Animals , Biological Transport , Host-Pathogen Interactions , Humans , Models, Biological , Nucleopolyhedroviruses/physiology , Vaccinia virus/physiology , Virus Diseases/virology
6.
PLoS Pathog ; 11(12): e1005342, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26700306

ABSTRACT

Orthopoxviruses (OPV), including variola, vaccinia, monkeypox, cowpox and ectromelia viruses cause acute infections in their hosts. With the exception of variola virus (VARV), the etiological agent of smallpox, other OPV have been reported to persist in a variety of animal species following natural or experimental infection. Despite the implications and significance for the ecology and epidemiology of diseases these viruses cause, those reports have never been thoroughly investigated. We used the mouse pathogen ectromelia virus (ECTV), the agent of mousepox and a close relative of VARV to investigate virus persistence in inbred mice. We provide evidence that ECTV causes a persistent infection in some susceptible strains of mice in which low levels of virus genomes were detected in various tissues late in infection. The bone marrow (BM) and blood appeared to be key sites of persistence. Contemporaneous with virus persistence, antiviral CD8 T cell responses were demonstrable over the entire 25-week study period, with a change in the immunodominance hierarchy evident during the first 3 weeks. Some virus-encoded host response modifiers were found to modulate virus persistence whereas host genes encoded by the NKC and MHC class I reduced the potential for persistence. When susceptible strains of mice that had apparently recovered from infection were subjected to sustained immunosuppression with cyclophosphamide (CTX), animals succumbed to mousepox with high titers of infectious virus in various organs. CTX treated index mice transmitted virus to, and caused disease in, co-housed naïve mice. The most surprising but significant finding was that immunosuppression of disease-resistant C57BL/6 mice several weeks after recovery from primary infection generated high titers of virus in multiple tissues. Resistant mice showed no evidence of a persistent infection. This is the strongest evidence that ECTV can persist in inbred mice, regardless of their resistance status.


Subject(s)
Ectromelia virus/immunology , Ectromelia, Infectious/immunology , Ectromelia, Infectious/transmission , Animals , Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recurrence
7.
Handb Exp Pharmacol ; 235: 237-261, 2017.
Article in English | MEDLINE | ID: mdl-27757755

ABSTRACT

The actin cytoskeleton is a crucial part of the eukaryotic cell. Viruses depend on host cells for their replication, and, as a result, many have developed ways of manipulating the actin network to promote their spread. This chapter reviews the various ways in which viruses utilize the actin cytoskeleton at discrete steps in their life cycle, from entry into the host cell, replication, and assembly of new progeny to virus release. Various actin inhibitors that function in different ways to affect proper actin dynamics can be used to parse the role of actin at these steps.


Subject(s)
Actin Cytoskeleton/physiology , Virus Replication , Animals , Clathrin/physiology , Host-Pathogen Interactions , Humans , Pinocytosis , Virus Assembly , Virus Internalization
8.
PLoS Pathog ; 9(3): e1003239, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555252

ABSTRACT

Cell-to-cell transmission of vaccinia virus can be mediated by enveloped virions that remain attached to the outer surface of the cell or those released into the medium. During egress, the outer membrane of the double-enveloped virus fuses with the plasma membrane leaving extracellular virus attached to the cell surface via viral envelope proteins. Here we report that F-actin nucleation by the viral protein A36 promotes the disengagement of virus attachment and release of enveloped virus. Cells infected with the A36(YdF) virus, which has mutations at two critical tyrosine residues abrogating localised actin nucleation, displayed a 10-fold reduction in virus release. We examined A36(YdF) infected cells by transmission electron microscopy and observed that during release, virus appeared trapped in small invaginations at the plasma membrane. To further characterise the mechanism by which actin nucleation drives the dissociation of enveloped virus from the cell surface, we examined recombinant viruses by super-resolution microscopy. Fluorescently-tagged A36 was visualised at sub-viral resolution to image cell-virus attachment in mutant and parental backgrounds. We confirmed that A36(YdF) extracellular virus remained closely associated to the plasma membrane in small membrane pits. Virus-induced actin nucleation reduced the extent of association, thereby promoting the untethering of virus from the cell surface. Virus release can be enhanced via a point mutation in the luminal region of B5 (P189S), another virus envelope protein. We found that the B5(P189S) mutation led to reduced contact between extracellular virus and the host membrane during release, even in the absence of virus-induced actin nucleation. Our results posit that during release virus is tightly tethered to the host cell through interactions mediated by viral envelope proteins. Untethering of virus into the surrounding extracellular space requires these interactions be relieved, either through the force of actin nucleation or by mutations in luminal proteins that weaken these interactions.


Subject(s)
Actin Cytoskeleton/metabolism , Vaccinia virus/physiology , Vaccinia/transmission , Viral Envelope Proteins/metabolism , Viral Structural Proteins/metabolism , Virus Release/physiology , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/ultrastructure , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Membrane/ultrastructure , Cell Membrane/virology , Chlorocebus aethiops , Comet Assay , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Fibroblasts/virology , Host-Pathogen Interactions , Mice , Microscopy, Electron, Transmission , NIH 3T3 Cells , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Vaccinia virus/ultrastructure , Vero Cells , Viral Envelope Proteins/ultrastructure , Viral Structural Proteins/ultrastructure
9.
Nature ; 458(7234): 87-91, 2009 Mar 05.
Article in English | MEDLINE | ID: mdl-19262673

ABSTRACT

Understanding cell motility will require detailed knowledge not only of the localization of signalling networks regulating actin polymerization, but also of their dynamics. Unfortunately, many signalling networks are not amenable to such analysis, as they are frequently transient and dispersed. By contrast, the signalling pathways used by pathogens undergoing actin-based motility are highly localized and operate in a constitutive fashion. Taking advantage of this, we have analysed the dynamics of neuronal Wiskott-Aldrich syndrome protein (N-WASP), WASP-interacting protein (WIP), GRB2 and NCK, which are required to stimulate actin-related protein (ARP)2/3-complex-dependent actin-based motility of vaccinia virus, using fluorescence recovery after photobleaching. Here we show that all four proteins are rapidly exchanging, albeit at different rates, and that the turnover of N-WASP depends on its ability to stimulate ARP2/3-complex-mediated actin polymerization. Conversely, disruption of the interaction of N-WASP with GRB2 and/or the barbed ends of actin filaments increases its exchange rate and results in a faster rate of virus movement. We suggest that the exchange rate of N-WASP controls the rate of ARP2/3-complex-dependent actin-based motility by regulating the extent of actin polymerization by antagonizing filament capping.


Subject(s)
Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Actins/chemistry , Adaptor Proteins, Signal Transducing , Biopolymers/chemistry , Biopolymers/metabolism , Cell Line , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Vaccinia virus/genetics , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics
10.
PLoS Pathog ; 8(6): e1002762, 2012.
Article in English | MEDLINE | ID: mdl-22685410

ABSTRACT

Paramount to the success of persistent viral infection is the ability of viruses to navigate hostile environments en route to future targets. In response to such obstacles, many viruses have developed the ability of establishing actin rich-membrane bridges to aid in future infections. Herein through dynamic imaging of HIV infected dendritic cells, we have observed how viral high-jacking of the actin/membrane network facilitates one of the most efficient forms of HIV spread. Within infected DC, viral egress is coupled to viral filopodia formation, with more than 90% of filopodia bearing immature HIV on their tips at extensions of 10 to 20 µm. Live imaging showed HIV filopodia routinely pivoting at their base, and projecting HIV virions at µm.sec⁻¹ along repetitive arc trajectories. HIV filopodial dynamics lead to up to 800 DC to CD4 T cell contacts per hour, with selection of T cells culminating in multiple filopodia tethering and converging to envelope the CD4 T-cell membrane with budding HIV particles. Long viral filopodial formation was dependent on the formin diaphanous 2 (Diaph2), and not a dominant Arp2/3 filopodial pathway often associated with pathogenic actin polymerization. Manipulation of HIV Nef reduced HIV transfer 25-fold by reducing viral filopodia frequency, supporting the potency of DC HIV transfer was dependent on viral filopodia abundance. Thus our observations show HIV corrupts DC to CD4 T cell interactions by physically embedding at the leading edge contacts of long DC filopodial networks.


Subject(s)
Carrier Proteins/metabolism , Dendritic Cells/virology , HIV , Host-Parasite Interactions/physiology , Pseudopodia/virology , CD4-Positive T-Lymphocytes/virology , Cell Line , Formins , Humans , Microscopy, Electron, Transmission , Pseudopodia/ultrastructure , Virion/metabolism , Virion/ultrastructure
11.
J Virol ; 86(13): 7427-43, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532690

ABSTRACT

Egress of wrapped virus (WV) to the cell periphery following vaccinia virus (VACV) replication is dependent on interactions with the microtubule motor complex kinesin-1 and is mediated by the viral envelope protein A36. Here we report that ectromelia virus (ECTV), a related orthopoxvirus and the causative agent of mousepox, encodes an A36 homologue (ECTV-Mos-142) that is highly conserved despite a large truncation at the C terminus. Deleting the ECTV A36R gene leads to a reduction in the number of extracellular viruses formed and to a reduced plaque size, consistent with a role in microtubule transport. We also observed a complete loss of virus-associated actin comets, another phenotype dependent on A36 expression during VACV infection. ECTV ΔA36R was severely attenuated when used to infect the normally susceptible BALB/c mouse strain. ECTV ΔA36R replication and spread from the draining lymph nodes to the liver and spleen were significantly reduced in BALB/c mice and in Rag-1-deficient mice, which lack T and B lymphocytes. The dramatic reduction in ECTV ΔA36R titers early during the course of infection was not associated with an augmented immune response. Taken together, these findings demonstrate the critical role that subcellular transport pathways play not only in orthopoxvirus infection in an in vitro context but also during orthopoxvirus pathogenesis in a natural host. Furthermore, despite the attenuation of the mutant virus, we found that infection nonetheless induced protective immunity in mice, suggesting that orthopoxvirus vectors with A36 deletions may be considered another safe vaccine alternative.


Subject(s)
Cytoskeletal Proteins/metabolism , Ectromelia virus/pathogenicity , Ectromelia, Infectious/virology , Host-Pathogen Interactions , Viral Proteins/metabolism , Virus Release , Animals , Ectromelia virus/genetics , Female , Gene Deletion , Liver/virology , Lymph Nodes/virology , Mice , Mice, Inbred BALB C , Protein Transport , Spleen/virology , Viral Load , Viral Plaque Assay , Viral Proteins/genetics , Virulence
12.
PLoS Genet ; 4(5): e1000085, 2008 May 30.
Article in English | MEDLINE | ID: mdl-18516287

ABSTRACT

Forward genetic screens in model organisms are an attractive means to identify those genes involved in any complex biological process, including neural circuit assembly. Although mutagenesis screens are readily performed to saturation, gene identification rarely is, being limited by the considerable effort generally required for positional cloning. Here, we apply a systematic positional cloning strategy to identify many of the genes required for neuronal wiring in the Drosophila visual system. From a large-scale forward genetic screen selecting for visual system wiring defects with a normal retinal pattern, we recovered 122 mutations in 42 genetic loci. For 6 of these loci, the underlying genetic lesions were previously identified using traditional methods. Using SNP-based mapping approaches, we have now identified 30 additional genes. Neuronal phenotypes have not previously been reported for 20 of these genes, and no mutant phenotype has been previously described for 5 genes. The genes encode a variety of proteins implicated in cellular processes such as gene regulation, cytoskeletal dynamics, axonal transport, and cell signalling. We conducted a comprehensive phenotypic analysis of 35 genes, scoring wiring defects according to 33 criteria. This work demonstrates the feasibility of combining large-scale gene identification with large-scale mutagenesis in Drosophila, and provides a comprehensive overview of the molecular mechanisms that regulate visual system wiring.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Nervous System/metabolism , Retina/metabolism , Animals , Axons/physiology , Cloning, Molecular , Drosophila/physiology , Mutation , Phenotype , Polymorphism, Single Nucleotide
13.
Dev Cell ; 56(4): 404-405, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33621490

ABSTRACT

Organisms resist bacterial infection at many levels. One of the least understood is the collective action of cells to limit pathogen spread. In this issue of Developmental Cell, Bastounis et al. (2021) describe the extrusion of Listeria monocytogenes from an epithelial monolayer by surrounding bystander cells.


Subject(s)
Listeria monocytogenes , Listeria , Humans
14.
Access Microbiol ; 3(3): 000206, 2021 Mar.
Article in English | MEDLINE | ID: mdl-34151161

ABSTRACT

HSV-1 envelope glycoprotein E (gE) is important for viral egress and cell-to-cell spread but the host protein(s) involved in these functions have yet to be determined. We aimed to investigate a role for the Arp2/3 complex and actin regulation in viral egress based on the identification of a WAVE Regulatory Complex (WRC) Interacting Receptor Sequence (WIRS) in the cytoplasmic tail (CT) of gE. A WIRS-dependent interaction between the gE(CT) and subunits of the WRC was demonstrated by GST-pulldown assay and a role for the Arp2/3 complex in cell-to-cell spread was also observed by plaque assay. Subsequent study of a recombinant HSV-1 gE WIRS-mutant found no significant changes to viral production and release based on growth kinetics studies, or changes to plaque and comet size in various cell types, suggesting no function for the motif in cell-to-cell spread. GFP-Trap pulldown and proximity ligation assays were unable to confirm a WIRS-dependent interaction between gE and the WRC in human cell lines though the WIRS-independent interaction observed in situ warrants further study. Confocal microscopy of infected cells of neuronal origin identified no impairment of gE WIRS-mutant HSV-1 anterograde transport along axons. We propose that the identified gE WIRS motif does not function directly in recruitment of the WRC in human cells, in cell-to-cell spread of virus or in anterograde transport along axons. Further studies are needed to understand how HSV-1 manipulates and traverses the actin cytoskeleton and how gE may contribute to these processes in a WIRS-independent manner.

15.
Cell Microbiol ; 11(5): 808-24, 2009 May.
Article in English | MEDLINE | ID: mdl-19207726

ABSTRACT

The vaccinia virus protein, F12, has been suggested to play an important role in microtubule-based transport of intracellular enveloped virus (IEV). We found that GFP-F12 is recruited to IEV moving on microtubules but is released from virus particles when they switch to actin-based motility. In the absence of F12, although the majority of IEV remain close to their peri-nuclear site of assembly, a small number of IEV still move with linear trajectories at speeds of 0.85 µm s(-1) , consistent with microtubule transport. Using a recombinant virus expressing GST-F12, we found that the viral protein E2 interacts directly with F12. In infected cells, GFP-E2 is observed on moving IEV as well as in the Golgi region, but is not associated with actin tails. In the absence of E2L, IEV accumulate in the peri-nuclear region and F12 is not recruited. Conversely, GFP-E2 is not observed on IEV in the absence of F12. Ultra-structural analysis of ΔE2L- and ΔF12L-infected cells reveals that loss of either protein results in defects in membrane wrapping during IEV formation. We suggest that E2 and F12 function as a complex that is necessary for IEV morphogenesis prior to their microtubule-based transport towards the plasma membrane.


Subject(s)
Vaccinia virus/physiology , Viral Proteins/metabolism , Cell Line , Green Fluorescent Proteins/analysis , Humans , Microtubules/metabolism , Protein Transport , Vaccinia virus/metabolism , Viral Proteins/analysis , Virion/metabolism
16.
Mol Biol Cell ; 18(1): 142-52, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17079733

ABSTRACT

Kinase D-interacting substrate of 220 kDa/ankyrin repeat-rich membrane spanning (Kidins220/ARMS) is a conserved membrane protein mainly expressed in brain and neuroendocrine cells, which is a downstream target of the signaling cascades initiated by neurotrophins and ephrins. We identified kinesin light chain 1 (KLC1) as a binding partner for Kidins220/ARMS by a yeast two-hybrid screen. The interaction between Kidins220/ARMS and the kinesin-1 motor complex was confirmed by glutathione S-transferase-pull-down and coimmunoprecipitation experiments. In addition, Kidins220/ARMS and kinesin-1 were shown to colocalize in nerve growth factor (NGF)-differentiated PC12 cells. Using Kidins220/ARMS and KLC1 mutants, we mapped the regions responsible for the binding to a short sequence of Kidins220/ARMS, termed KLC-interacting motif (KIM), which is sufficient for the interaction with KLC1. Optimal binding of KIM requires a region of KLC1 spanning both the tetratricopeptide repeats and the heptad repeats, previously not involved in cargo recognition. Overexpression of KIM in differentiating PC12 cells impairs the formation and transport of EGFP-Kidins220/ARMS carriers to the tips of growing neurites, leaving other kinesin-1 dependent processes unaffected. Furthermore, KIM overexpression interferes with the activation of the mitogen-activated protein kinase signaling and neurite outgrowth in NGF-treated PC12 cells. Our results suggest that Kidins220/ARMS-positive carriers undergo a kinesin-1-dependent transport linked to neurotrophin action.


Subject(s)
Cell Differentiation , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Phosphoproteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Differentiation/drug effects , Gene Expression/drug effects , HeLa Cells , Humans , Kinesins , Membrane Proteins/chemistry , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Nerve Growth Factor/pharmacology , Neurons/drug effects , PC12 Cells , Phosphoproteins/chemistry , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Structure, Tertiary/drug effects , Protein Transport/drug effects , Rats
17.
J Med Virol ; 81(9): 1674-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19626609

ABSTRACT

There is a lack of quantitative information about the generation of virus aerosols by infected subjects. The exhaled aerosols generated by coughing, talking, and breathing were sampled in 50 subjects using a novel mask, and analyzed using PCR for nine respiratory viruses. The exhaled samples from a subset of 10 subjects who were PCR positive for rhinovirus were also examined by cell culture for this virus. Of the 50 subjects, among the 33 with symptoms of upper respiratory tract infections, 21 had at least one virus detected by PCR, while amongst the 17 asymptomatic subjects, 4 had a virus detected by PCR. Overall, rhinovirus was detected in 19 subjects, influenza in 4 subjects, parainfluenza in 2 subjects, and human metapneumovirus in 1 subject. Two subjects were co-infected. Of the 25 subjects who had virus-positive nasal mucus, the same virus type was detected in 12 breathing samples, 8 talking samples, and in 2 coughing samples. In the subset of exhaled samples from 10 subjects examined by culture, infective rhinovirus was detected in 2. These data provide further evidence that breathing may be a source of respirable particles carrying infectious virus.


Subject(s)
Cough , Exhalation , Respiration , Virus Shedding , Viruses/classification , Viruses/isolation & purification , Adult , Aerosols , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Polymerase Chain Reaction/methods , Young Adult
18.
Methods Mol Biol ; 2023: 73-92, 2019.
Article in English | MEDLINE | ID: mdl-31240671

ABSTRACT

Vaccinia virus recombinants that express fluorescent proteins have a variety of applications such as the identification of infected cells, efficient screening for genetically modified strains, and molecular characterization of virus replication and spread. The detection of fluorescent proteins and viral-fluorescent fusion proteins by fluorescence microscopy is noninvasive and can be used to describe protein localization in live cells and track the intracellular movement of virus particles. This chapter describes a number of approaches for the construction of plasmids and subsequent generation and isolation of fluorescent recombinant viruses.


Subject(s)
Luminescent Proteins/biosynthesis , Microscopy, Fluorescence/methods , Vaccinia virus/genetics , Plasmids/genetics , Virion/genetics
19.
Viruses ; 10(8)2018 07 24.
Article in English | MEDLINE | ID: mdl-30042325

ABSTRACT

The microtubule cytoskeleton is a primary organizer of viral infections for delivering virus particles to their sites of replication, establishing and maintaining subcellular compartments where distinct steps of viral morphogenesis take place, and ultimately dispersing viral progeny. One of the best characterized examples of virus motility is the anterograde transport of the wrapped virus form of vaccinia virus (VACV) from the trans-Golgi network (TGN) to the cell periphery by kinesin-1. Yet many aspects of this transport event are elusive due to the speed of motility and the challenges of imaging this stage at high resolution over extended time periods. We have established a novel imaging technology to track virus transport that uses photoconvertible fluorescent recombinant viruses to track subsets of virus particles from their site of origin and determine their destination. Here we image virus exit from the TGN and their rate of egress to the cell periphery. We demonstrate a role for kinesin-1 engagement in regulating virus exit from the TGN by removing A36 and F12 function, critical viral mediators of kinesin-1 recruitment to virus particles. Phototracking viral particles and components during infection is a powerful new imaging approach to elucidate mechanisms of virus replication.


Subject(s)
Cytoplasm/metabolism , Vaccinia virus/physiology , Virion/physiology , Virus Release , trans-Golgi Network/physiology , Biological Transport , Cytoplasm/virology , HeLa Cells , Humans , Kinesins/metabolism , Optical Imaging , Vaccinia virus/genetics , Viral Proteins/genetics , Viral Structural Proteins/genetics , Virus Replication , trans-Golgi Network/virology
20.
Viruses ; 10(3)2018 03 05.
Article in English | MEDLINE | ID: mdl-29510577

ABSTRACT

Ectromelia virus (ECTV) is an orthopoxvirus and the causative agent of mousepox. Like other poxviruses such as variola virus (agent of smallpox), monkeypox virus and vaccinia virus (the live vaccine for smallpox), ECTV promotes actin-nucleation at the surface of infected cells during virus release. Homologs of the viral protein A36 mediate this function through phosphorylation of one or two tyrosine residues that ultimately recruit the cellular Arp2/3 actin-nucleating complex. A36 also functions in the intracellular trafficking of virus mediated by kinesin-1. Here, we describe the generation of a recombinant ECTV that is specifically disrupted in actin-based motility allowing us to examine the role of this transport step in vivo for the first time. We show that actin-based motility has a critical role in promoting the release of virus from infected cells in vitro but plays a minor role in virus spread in vivo. It is likely that loss of microtubule-dependent transport is a major factor for the attenuation observed when A36R is deleted.


Subject(s)
Actins/metabolism , Ectromelia virus/physiology , Ectromelia, Infectious/metabolism , Ectromelia, Infectious/virology , Virus Release , Animals , Biological Transport , Cell Line , Chlorocebus aethiops , Gene Expression , Humans , Mice , Mice, Knockout , Mutation , Protein Binding , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL