Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Gen Virol ; 103(1)2022 01.
Article in English | MEDLINE | ID: mdl-35020582

ABSTRACT

The morphogenesis of vaccinia virus (VACV, family Poxviridae), the smallpox vaccine, is a complex process involving multiple distinct cellular membranes and resulting in multiple different forms of infectious virion. Efficient release of enveloped virions, which promote systemic spread of infection within hosts, requires the VACV protein E2 but the molecular basis of E2 function remains unclear and E2 lacks sequence homology to any well-characterised family of proteins. We solved the crystal structure of VACV E2 to 2.3 Å resolution, revealing that it comprises two domains with novel folds: an N-terminal annular (ring) domain and a C-terminal globular (head) domain. The C-terminal head domain displays weak structural homology with cellular (pseudo)kinases but lacks conserved surface residues or kinase features, suggesting that it is not enzymatically active, and possesses a large surface basic patch that might interact with phosphoinositide lipid headgroups. Recent deep learning methods have revolutionised our ability to predict the three-dimensional structures of proteins from primary sequence alone. VACV E2 is an exemplar 'difficult' viral protein target for structure prediction, being comprised of multiple novel domains and lacking sequence homologues outside Poxviridae. AlphaFold2 nonetheless succeeds in predicting the structures of the head and ring domains with high and moderate accuracy, respectively, allowing accurate inference of multiple structural properties. The advent of highly accurate virus structure prediction marks a step-change in structural virology and beckons a new era of structurally-informed molecular virology.


Subject(s)
Poxviridae/metabolism , Vaccinia virus/chemistry , Vaccinia virus/physiology , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Replication , Binding Sites , Crystallography, X-Ray , Protein Binding , Protein Conformation , Vaccinia virus/genetics , Viral Proteins/genetics
2.
J Gen Virol ; 98(10): 2543-2555, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28933687

ABSTRACT

Vaccinia virus produces two distinct infectious virions; the single-enveloped intracellular mature virus (IMV), which remains in the cell until cell lysis, and the double-enveloped extracellular enveloped virus (EEV), which mediates virus spread. The latter is derived from a triple-enveloped intracellular enveloped virus (IEV) precursor, which is transported to the cell periphery by the kinesin-1 motor complex. This transport involves the viral protein A36 as well as F12 and E2. A36 is an integral membrane protein associated with the outer virus envelope and is the only known direct link between virion and kinesin-1 complex. Yet in the absence of A36 virion egress still occurs on microtubules, albeit at reduced efficiency. In this paper double-fluorescent labelling of the capsid protein A5 and outer-envelope protein F13 was exploited to visualize IEV transport by live-cell imaging in the absence of either A36 or F12. During the generation of recombinant viruses expressing both A5-GFP and F13-mCherry a plaque size defect was identified that was particularly severe in viruses lacking A36. Electron microscopy showed that this phenotype was caused by abnormal wrapping of IMV to form IEV, and this resulted in reduced virus egress to the cell surface. The aberrant wrapping phenotype suggests that the fluorescent fusion protein interferes with an interaction of F13 with the IMV surface that is required for tight association between IMVs and wrapping membranes. The severity of this defect suggests that these viruses are imperfect tools for characterizing virus egress.

3.
J Gen Virol ; 98(6): 1500-1514, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28631604

ABSTRACT

Egress of vaccinia virus from its host cell is mediated by the microtubule-associated motor kinesin-1, and three viral proteins, A36 and the F12/E2 complex, have been implicated in this process. Deletion of F12 expression causes a more severe reduction in egress than deletion of A36 but whether these proteins are involved in the same or different mechanisms of kinesin-1 recruitment is unknown. Here it is shown that a virus lacking both proteins forms a smaller plaque than mutants lacking either gene alone, indicating non-redundant functions. A36 not only links virions directly to kinesin-1 but also nucleates actin polymerization to propel surface virions away from the host cell. To address the relative importance of these functions for virus spread, a panel of recombinant viruses was constructed in which the ability of A36 to bind kinesin-1 or to nucleate actin polymerization was abrogated individually or together, in the presence or absence of F12 expression. Analysis of these viruses revealed that in the presence of the F12 protein, loss of kinesin-1 interaction made a greater contribution to plaque size than did the formation of actin tails. However in the absence of F12, the ability of A36 to promote egress was abrogated. Therefore, the ability of A36 to promote egress by kinesin-1 is reliant on the F12 protein.


Subject(s)
Vaccinia virus/physiology , Viral Proteins/metabolism , Virus Release , Animals , Cell Line , Gene Deletion , Host-Pathogen Interactions , Humans , Kinesins/metabolism , Protein Interaction Mapping , Viral Plaque Assay , Viral Proteins/genetics
4.
Traffic ; 18(8): 505-518, 2017 08.
Article in English | MEDLINE | ID: mdl-28485852

ABSTRACT

Vaccinia virus (VACV) utilizes microtubule-mediated trafficking at several stages of its life cycle, of which virus egress is the most intensely studied. During egress VACV proteins A36, F12 and E2 are involved in kinesin-1 interactions; however, the roles of these proteins remain poorly understood. A36 forms a direct link between virions and kinesin-1, yet in its absence VACV egress still occurs on microtubules. During a co-immunoprecipitation screen to seek an alternative link between virions and kinesin, A36 was found to bind isoform KLC1 rather than KLC2. The F12/E2 complex associates preferentially with the C-terminal tail of KLC2, to a region that overlaps the binding site of cellular 14-3-3 proteins. F12/E2 displaces 14-3-3 from KLC and, unlike 14-3-3, does not require phosphorylation of KLC for its binding. The region determining the KLC1 specificity of A36 was mapped to the KLC N-terminal heptad repeat region that is responsible for its association with kinesin heavy chain. Despite these differing binding properties F12/E2 can co-operatively enhance A36 association with KLC, particularly when using a KLC1-KLC2 chimaera that resembles several KLC1 spliceforms and can bind A36 and F12/E2 efficiently. This is the first example of a pathogen encoding multiple proteins that co-operatively associate with kinesin-1.


Subject(s)
Kinesins/metabolism , Protein Isoforms/metabolism , Vaccinia virus/metabolism , Viral Proteins/metabolism , 14-3-3 Proteins/metabolism , Animals , Cell Line , Humans , Protein Binding , Protein Transport
5.
PLoS Pathog ; 11(3): e1004723, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25760349

ABSTRACT

During vaccinia virus morphogenesis, intracellular mature virus (IMV) particles are wrapped by a double lipid bilayer to form triple enveloped virions called intracellular enveloped virus (IEV). IEV are then transported to the cell surface where the outer IEV membrane fuses with the cell membrane to expose a double enveloped virion outside the cell. The F12, E2 and A36 proteins are involved in transport of IEVs to the cell surface. Deletion of the F12L or E2L genes causes a severe inhibition of IEV transport and a tiny plaque size. Deletion of the A36R gene leads to a smaller reduction in plaque size and less severe inhibition of IEV egress. The A36 protein is present in the outer membrane of IEVs, and over-expressed fragments of this protein interact with kinesin light chain (KLC). However, no interaction of F12 or E2 with the kinesin complex has been reported hitherto. Here the F12/E2 complex is shown to associate with kinesin-1 through an interaction of E2 with the C-terminal tail of KLC isoform 2, which varies considerably between different KLC isoforms. siRNA-mediated knockdown of KLC isoform 1 increased IEV transport to the cell surface and virus plaque size, suggesting interaction with KLC isoform 1 is somehow inhibitory of IEV transport. In contrast, knockdown of KLC isoform 2 did not affect IEV egress or plaque formation, indicating redundancy in virion egress pathways. Lastly, the enhancement of plaque size resulting from loss of KLC isoform 1 was abrogated by removal of KLC isoforms 1 and 2 simultaneously. These observations suggest redundancy in the mechanisms used for IEV egress, with involvement of KLC isoforms 1 and 2, and provide evidence of interaction of F12/E2 complex with the kinesin-1 complex.


Subject(s)
Microtubule-Associated Proteins/metabolism , Vaccinia virus/metabolism , Vaccinia virus/pathogenicity , Viral Proteins/metabolism , Flow Cytometry , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Kinesins , Microscopy, Confocal , Protein Transport/physiology , Transfection
6.
J Biol Chem ; 289(23): 16148-63, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24753246

ABSTRACT

Rabies pseudotyped lentiviral vectors have great potential in gene therapy, not least because of their ability to transduce neurons following their distal axonal application. However, very little is known about the molecular processes that underlie their retrograde transport and cell transduction. Using multiple labeling techniques and confocal microscopy, we demonstrated that pseudotyping with rabies virus envelope glycoprotein (RV-G) enabled the axonal retrograde transport of two distinct subtypes of lentiviral vector in motor neuron cultures. Analysis of this process revealed that these vectors trafficked through Rab5-positive endosomes and accumulated within a non-acidic Rab7 compartment. RV-G pseudotyped vectors were co-transported with both the tetanus neurotoxin-binding fragment and the membrane proteins thought to mediate rabies virus endocytosis (neural cell adhesion molecule, nicotinic acetylcholine receptor, and p75 neurotrophin receptor), thus demonstrating that pseudotyping with RV-G targets lentiviral vectors for transport along the same pathway exploited by several toxins and viruses. Using motor neurons cultured in compartmentalized chambers, we demonstrated that axonal retrograde transport of these vectors was rapid and efficient; however, it was not able to transduce the targeted neurons efficiently, suggesting that impairment in processes occurring after arrival of the viral vector in the soma is responsible for the low transduction efficiency seen in vivo, which suggests a novel area for improvement of gene therapy vectors.


Subject(s)
Axonal Transport , Genetic Vectors , Lentivirus/genetics , Motor Neurons/metabolism , Rabies virus/metabolism , Viral Envelope Proteins/metabolism , Animals , Endocytosis , HEK293 Cells , Humans , Motor Neurons/virology , Rats , Viral Envelope Proteins/genetics
7.
J Gen Virol ; 94(Pt 4): 720-725, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23255618

ABSTRACT

Vaccinia virus (VACV) has two infectious forms called intracellular mature virus and extracellular enveloped virus (EEV). Two of the seven viral proteins in the EEV outer envelope, A33 and A34, are type II membrane glycoproteins that each interact with another EEV protein called B5; however, evidence for direct A33-A34 interaction is lacking. The localization and stability of A34 is affected by B5 and here data are presented showing that A34 is also affected by A33. In the absence of A33, just as without B5, the level, localization and glycosylation profile of A34 was altered. However, the glycosylation profile of A34 without A33 is different to that observed in the absence of B5, and A34 accumulates in the Golgi apparatus rather than in the endoplasmic reticulum. Thus, A34 requires more than one other EEV protein for its processing and cellular transport.


Subject(s)
Glycoproteins/metabolism , Membrane Glycoproteins/metabolism , Protein Interaction Mapping , Vaccinia virus/physiology , Viral Envelope Proteins/metabolism , Virus Replication , Protein Stability , Protein Transport
8.
Biotechnol Appl Biochem ; 56(3): 85-93, 2010 Jun 21.
Article in English | MEDLINE | ID: mdl-20441568

ABSTRACT

Secretory and membrane-bound proteins are generally produced in lower amounts in insect cells compared with cytoplasmic and nuclear proteins. There may be many reasons for this, including degradation of recombinant proteins by proteases, competition for cellular resources between native and recombinant proteins, and physical blockage of the secretory pathways. In the present study, we describe the construction of a baculovirus in which chiA (chitinase) and cath (cathepsin) genes have been deleted and show improved recombinant protein expression using this vector. We confirmed the complete removal of both genes by PCR, restriction enzyme analysis and enzyme assays, and the modified virus DNA was shown to be stable in bacterial cells over multiple passages. A selection of recombinant genes were inserted into the double-deletion virus and their expression levels compared with recombinant viruses that had single or no gene deletions. In all instances, the double-deletion viruses showed greatly enhanced levels of protein production for both secreted and nuclear/cytoplasmic proteins. In summary, we have conclusively demonstrated the importance of this deletion vector for the high-level production of recombinant proteins.


Subject(s)
Baculoviridae/genetics , Membrane Proteins/biosynthesis , Protein Engineering/methods , Recombinant Proteins/biosynthesis , Animals , Baculoviridae/enzymology , Cathepsins/genetics , Cells, Cultured , Chitinases/genetics , Gene Deletion , Gene Expression , Humans , Insecta/cytology , Membrane Proteins/genetics
9.
PLoS Pathog ; 6(2): e1000785, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20195521

ABSTRACT

Vaccinia virus (VACV) uses microtubules for export of virions to the cell surface and this process requires the viral protein F12. Here we show that F12 has structural similarity to kinesin light chain (KLC), a subunit of the kinesin-1 motor that binds cargo. F12 and KLC share similar size, pI, hydropathy and cargo-binding tetratricopeptide repeats (TPRs). Moreover, molecular modeling of F12 TPRs upon the crystal structure of KLC2 TPRs showed a striking conservation of structure. We also identified multiple TPRs in VACV proteins E2 and A36. Data presented demonstrate that F12 is critical for recruitment of kinesin-1 to virions and that a conserved tryptophan and aspartic acid (WD) motif, which is conserved in the kinesin-1-binding sequence (KBS) of the neuronal protein calsyntenin/alcadein and several other cellular kinesin-1 binding proteins, is essential for kinesin-1 recruitment and virion transport. In contrast, mutation of WD motifs in protein A36 revealed they were not required for kinesin-1 recruitment or IEV transport. This report of a viral KLC-like protein containing a KBS that is conserved in several cellular proteins advances our understanding of how VACV recruits the kinesin motor to virions, and exemplifies how viruses use molecular mimicry of cellular components to their advantage.


Subject(s)
Microtubule-Associated Proteins/chemistry , Vaccinia virus/physiology , Viral Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Conserved Sequence , Cryoelectron Microscopy , HeLa Cells , Humans , Kinesins , Microscopy, Immunoelectron , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Models, Molecular , Molecular Mimicry , Molecular Sequence Data , Polymerase Chain Reaction , Protein Structure, Tertiary , Vaccinia virus/pathogenicity , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/pathogenicity , Virion/physiology
10.
Virology ; 371(2): 278-91, 2008 Feb 20.
Article in English | MEDLINE | ID: mdl-17991504

ABSTRACT

The role of the microtubule-associated P10 protein of baculoviruses is not yet understood. P10 has previously been linked with the formation of a number of cytoskeletal-like or cytoskeleton-associated structures in the nucleus and cytoplasm, thought to be involved in the morphogenesis of virus polyhedral occlusion bodies. The formation of these structures was studied by immunofluorescence laser scanning confocal microscopy in TN368 cells, a model system amenable to the study of virus interaction with the host cell cytoskeleton. We show that the Autographa californica nucleopolyhedrovirus P10 protein forms two distinct cytoskeletal-like structures, microtubule-associated filaments and perinuclear tubular aggregates. P10 also associates with polyhedral occlusion bodies. Depolymerisation of the microtubule network with colchicine prevents formation of P10 filaments but not of P10 tubules. Colchicine treatment enhances the association of P10 with occlusion bodies. Transient expression of P10 showed that both filaments and tubules can form in the absence of other viral proteins. We postulate a number of possible roles of the P10 protein during virus infection and morphogenesis.


Subject(s)
Cytoskeleton/metabolism , Inclusion Bodies, Viral/metabolism , Lepidoptera/virology , Nucleopolyhedroviruses/pathogenicity , Viral Proteins/metabolism , Animals , Cells, Cultured , Computational Biology , Cytoskeleton/ultrastructure , Inclusion Bodies, Viral/ultrastructure , Microscopy, Confocal , Microscopy, Fluorescence , Microtubules/metabolism , Microtubules/ultrastructure , Nucleopolyhedroviruses/metabolism , Nucleopolyhedroviruses/ultrastructure , Spodoptera/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...