Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Viruses ; 16(4)2024 04 03.
Article in English | MEDLINE | ID: mdl-38675899

ABSTRACT

Lumpy skin disease virus (LSDV) is a member of the capripoxvirus (CPPV) genus of the Poxviridae family. LSDV is a rapidly emerging, high-consequence pathogen of cattle, recently spreading from Africa and the Middle East into Europe and Asia. We have sequenced the whole genome of historical LSDV isolates from the Pirbright Institute virus archive, and field isolates from recent disease outbreaks in Sri Lanka, Mongolia, Nigeria and Ethiopia. These genome sequences were compared to published genomes and classified into different subgroups. Two subgroups contained vaccine or vaccine-like samples ("Neethling-like" clade 1.1 and "Kenya-like" subgroup, clade 1.2.2). One subgroup was associated with outbreaks of LSD in the Middle East/Europe (clade 1.2.1) and a previously unreported subgroup originated from cases of LSD in west and central Africa (clade 1.2.3). Isolates were also identified that contained a mix of genes from both wildtype and vaccine samples (vaccine-like recombinants, grouped in clade 2). Whole genome sequencing and analysis of LSDV strains isolated from different regions of Africa, Europe and Asia have provided new knowledge of the drivers of LSDV emergence, and will inform future disease control strategies.


Subject(s)
Genome, Viral , Lumpy Skin Disease , Lumpy skin disease virus , Phylogeny , Whole Genome Sequencing , Lumpy skin disease virus/genetics , Lumpy skin disease virus/classification , Lumpy skin disease virus/isolation & purification , Animals , Lumpy Skin Disease/virology , Lumpy Skin Disease/epidemiology , Cattle , Africa, Central/epidemiology , Africa, Western/epidemiology , Disease Outbreaks
2.
Front Immunol ; 13: 1051008, 2022.
Article in English | MEDLINE | ID: mdl-36518761

ABSTRACT

Lumpy skin disease virus (LSDV) causes severe disease in cattle and water buffalo and is transmitted by hematophagous arthropod vectors. Detailed information of the adaptive and innate immune response to LSDV is limited, hampering the development of tools to control the disease. This study provides an in-depth analysis of the immune responses of calves experimentally inoculated with LSDV via either needle-inoculation or arthropod-inoculation using virus-positive Stomoxys calcitrans and Aedes aegypti vectors. Seven out of seventeen needle-inoculated calves (41%) developed clinical disease characterised by multifocal necrotic cutaneous nodules. In comparison 8/10 (80%) of the arthropod-inoculated calves developed clinical disease. A variable LSDV-specific IFN-γ immune response was detected in the needle-inoculated calves from 5 days post inoculation (dpi) onwards, with no difference between clinical calves (developed cutaneous lesions) and nonclinical calves (did not develop cutaneous lesions). In contrast a robust and uniform cell-mediated immune response was detected in all eight clinical arthropod-inoculated calves, with little response detected in the two nonclinical arthropod-inoculated calves. Neutralising antibodies against LSDV were detected in all inoculated cattle from 5-7 dpi. Comparison of the production of anti-LSDV IgM and IgG antibodies revealed no difference between clinical and nonclinical needle-inoculated calves, however a strong IgM response was evident in the nonclinical arthropod-inoculated calves but absent in the clinical arthropod-inoculated calves. This suggests that early IgM production is a correlate of protection in LSD. This study presents the first evidence of differences in the immune response between clinical and nonclinical cattle and highlights the importance of using a relevant transmission model when studying LSD.


Subject(s)
Lumpy Skin Disease , Lumpy skin disease virus , Cattle , Animals , Lumpy skin disease virus/physiology , Lumpy Skin Disease/prevention & control , Mosquito Vectors , Immunity, Cellular , Buffaloes , Immunoglobulin M
3.
J Virol ; 96(15): e0075122, 2022 08 10.
Article in English | MEDLINE | ID: mdl-35867566

ABSTRACT

Lumpy skin disease virus (LSDV) is a poxvirus that causes severe systemic disease in cattle and is spread by mechanical arthropod-borne transmission. This study quantified the acquisition and retention of LSDV by four species of Diptera (Stomoxys calcitrans, Aedes aegypti, Culex quinquefasciatus, and Culicoides nubeculosus) from cutaneous lesions, normal skin, and blood from a clinically affected animal. The acquisition and retention of LSDV by Ae. aegypti from an artificial membrane feeding system was also examined. Mathematical models of the data were generated to identify the parameters which influence insect acquisition and retention of LSDV. For all four insect species, the probability of acquiring LSDV was substantially greater when feeding on a lesion compared with feeding on normal skin or blood from a clinically affected animal. After feeding on a skin lesion LSDV was retained on the proboscis for a similar length of time (around 9 days) for all four species and for a shorter time in the rest of the body, ranging from 2.2 to 6.4 days. Acquisition and retention of LSDV by Ae. aegypti after feeding on an artificial membrane feeding system that contained a high titer of LSDV was comparable to feeding on a skin lesion on a clinically affected animal, supporting the use of this laboratory model as a replacement for some animal studies. This work reveals that the cutaneous lesions of LSD provide the high-titer source required for acquisition of the virus by insects, thereby enabling the mechanical vector-borne transmission. IMPORTANCE Lumpy skin disease virus (LSDV) is a high consequence pathogen of cattle that is rapidly expanding its geographical boundaries into new regions such as Europe and Asia. This expansion is promoted by the mechanical transmission of the virus via hematogenous arthropods. This study quantifies the acquisition and retention of LSDV by four species of blood-feeding insects and reveals that the cutaneous lesions of LSD provide the high titer virus source necessary for virus acquisition by the insects. An artificial membrane feeding system containing a high titer of LSDV was shown to be comparable to a skin lesion on a clinically affected animal when used as a virus source. This promotes the use of these laboratory-based systems as replacements for some animal studies. Overall, this work advances our understanding of the mechanical vector-borne transmission of LSDV and provides evidence to support the design of more effective disease control programmes.


Subject(s)
Blood , Diptera , Feeding Behavior , Insect Vectors , Lumpy Skin Disease , Lumpy skin disease virus , Aedes/anatomy & histology , Aedes/virology , Animals , Cattle/virology , Ceratopogonidae/anatomy & histology , Ceratopogonidae/virology , Culex/anatomy & histology , Culex/virology , Diptera/anatomy & histology , Diptera/physiology , Diptera/virology , Insect Vectors/anatomy & histology , Insect Vectors/physiology , Insect Vectors/virology , Lumpy Skin Disease/virology , Lumpy skin disease virus/isolation & purification , Lumpy skin disease virus/physiology , Membranes, Artificial , Muscidae/anatomy & histology , Muscidae/virology , Time Factors
4.
Transbound Emerg Dis ; 69(4): e336-e343, 2022 Jul.
Article in English | MEDLINE | ID: mdl-34448540

ABSTRACT

Lumpy skin disease virus (LSDV) is an emerging poxviral pathogen of cattle that is currently spreading throughout Asia. The disease situation is of high importance for farmers and policy makers in Asia. In October 2020, feral cattle in Hong Kong developed multi-focal cutaneous nodules consistent with lumpy skin disease (LSD). Gross and histological pathology further supported the diagnosis and samples were sent to the OIE Reference Laboratory at The Pirbright Institute for confirmatory testing. LSDV was detected using quantitative polymerase chain reaction (qPCR) and additional molecular analyses. This is the first report of LSD in Hong Kong. Whole genome sequencing (WGS) of the strain LSDV/Hong Kong/2020 and phylogenetic analysis were carried out in order to identify connections to previous outbreaks of LSD, and better understand the drivers of LSDV emergence. Analysis of the 90 core poxvirus genes revealed LSDV/Hong Kong/2020 was a novel strain most closely related to the live-attenuated Neethling vaccine strains of LSDV and more distantly related to wildtype LSDV isolates from Africa, the Middle East and Europe. Analysis of the more variable regions located towards the termini of the poxvirus genome revealed genes in LSDV/Hong Kong/2020 with different patterns of grouping when compared to previously published wildtype and vaccine strains of LSDV. This work reveals that the LSD outbreak in Hong Kong in 2020 was caused by a different strain of LSDV than the LSD epidemic in the Middle East and Europe in 2015-2018. The use of WGS is highly recommended when investigating LSDV disease outbreaks.


Subject(s)
Cattle Diseases , Lumpy Skin Disease , Lumpy skin disease virus , Animals , Cattle , Cattle Diseases/epidemiology , Disease Outbreaks/veterinary , Hong Kong/epidemiology , Phylogeny , Vaccines, Attenuated
5.
J Virol ; 95(9)2021 04 12.
Article in English | MEDLINE | ID: mdl-33568514

ABSTRACT

Lumpy skin disease virus (LSDV) is a vector-transmitted poxvirus that causes disease in cattle. Vector species involved in LSDV transmission and their ability to acquire and transmit the virus are poorly characterized. Using a highly representative bovine experimental model of lumpy skin disease, we fed four model vector species (Aedes aegypti, Culex quinquefasciatus, Stomoxys calcitrans, and Culicoides nubeculosus) on LSDV-inoculated cattle in order to examine their acquisition and retention of LSDV. Subclinical disease was a more common outcome than clinical disease in the inoculated cattle. Importantly, the probability of vectors acquiring LSDV from a subclinical animal (0.006) was very low compared with that from a clinical animal (0.23), meaning an insect feeding on a subclinical animal was 97% less likely to acquire LSDV than one feeding on a clinical animal. All four potential vector species studied acquired LSDV from the host at a similar rate, but Aedes aegypti and Stomoxys calcitrans retained the virus for a longer time, up to 8 days. There was no evidence of virus replication in the vector, consistent with mechanical rather than biological transmission. The parameters obtained in this study were combined with data from studies of LSDV transmission and vector life history parameters to determine the basic reproduction number of LSDV in cattle mediated by each of the model species. This reproduction number was highest for Stomoxys calcitrans (19.1), followed by C. nubeculosus (7.1) and Ae. aegypti (2.4), indicating that these three species are potentially efficient transmitters of LSDV; this information can be used to inform LSD control programs.IMPORTANCE Lumpy skin disease virus (LSDV) causes a severe systemic disease characterized by cutaneous nodules in cattle. LSDV is a rapidly emerging pathogen, having spread since 2012 into Europe and Russia and across Asia. The vector-borne nature of LSDV transmission is believed to have promoted this rapid geographic spread of the virus; however, a lack of quantitative evidence about LSDV transmission has hampered effective control of the disease during the current epidemic. Our research shows subclinical cattle play little part in virus transmission relative to clinical cattle and reveals a low probability of virus acquisition by insects at the preclinical stage. We have also calculated the reproductive number of different insect species, therefore identifying efficient transmitters of LSDV. This information is of utmost importance, as it will help to define epidemiological control measures during LSDV epidemics and of particular consequence in resource-poor regions where LSD vaccination may be less than adequate.


Subject(s)
Insect Vectors , Lumpy Skin Disease/transmission , Lumpy skin disease virus/physiology , Animals , Cattle , Insect Vectors/physiology , Insect Vectors/virology , Male , Virus Replication
6.
J Virol Methods ; 285: 113943, 2020 11.
Article in English | MEDLINE | ID: mdl-32707050

ABSTRACT

Lumpy skin disease virus (LSDV) is a poxvirus that causes systemic disease in cattle, resulting in substantial economic loss to affected communities. LSDV is a rapidly emerging pathogen of growing global concern that recently spread from Africa and the Middle East into Europe and Asia, impacting the cattle population in these regions. An increase in research efforts into LSDV is required to address key knowledge gaps, however this is hampered by lack of suitable cell lines on which to propagate and study the virus. In this work we describe the replication and spread of LSDV on Madin-Darby bovine kidney (MDBK) cells, and the formation of foci-type poxvirus plaques by LSDV on MDBK cells. Methods utilising MDBK cells to quantify neutralising antibodies to LSDV, and to purify LSDV genomic DNA suitable for short read sequencing are described. These research methods broaden the tools available for LSDV researchers and will facilitate the gathering of evidence to underpin the development of LSD control and prevention programmes.


Subject(s)
DNA, Viral/isolation & purification , Lumpy Skin Disease/virology , Lumpy skin disease virus , Virus Cultivation , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cattle , Dogs , Lumpy skin disease virus/isolation & purification , Lumpy skin disease virus/physiology , Madin Darby Canine Kidney Cells
7.
Vet Pathol ; 57(3): 388-396, 2020 05.
Article in English | MEDLINE | ID: mdl-32314676

ABSTRACT

Lumpy skin disease is a high-consequence disease in cattle caused by infection with the poxvirus lumpy skin disease virus (LSDV). The virus is endemic in most countries in Africa and an emerging threat to cattle populations in Europe and Asia. As LSDV spreads into new regions, it is important that signs of disease are recognized promptly by animal caregivers. This study describes the gross, microscopic, and ultrastructural changes that occur over time in cattle experimentally challenged with LSDV. Four calves were inoculated with wildtype LSDV and monitored for 19 to 21 days. At 7 days after inoculation, 2 of the 4 cattle developed multifocal cutaneous nodules characteristic of LSD. Some lesions displayed a targetoid appearance. Histologically, intercellular and intracellular edema was present in the epidermis of some nodules. Occasional intracytoplasmic inclusion bodies were identified in keratinocytes. More severe and consistent changes were present in the dermis, with marked histiocytic inflammation and necrotizing fibrinoid vasculitis of dermal vessels, particularly the deep dermal plexus. Chronic lesions consisted of full-thickness necrosis of the dermis and epidermis. Lesions in other body organs were not a major feature of LSD in this study, highlighting the strong cutaneous tropism of this virus. Immunohistochemistry and electron microscopy identified LSDV-infected histiocytes and fibroblasts in the skin nodules of affected cattle. This study highlights the noteworthy lesions of LSDV and how they develop over time.


Subject(s)
Lumpy Skin Disease , Lumpy skin disease virus/isolation & purification , Animals , Asia/epidemiology , Cattle , Cattle Diseases/virology , Communicable Diseases, Emerging/veterinary , Communicable Diseases, Emerging/virology , Dermatitis/pathology , Dermatitis/veterinary , Dermatitis/virology , Endemic Diseases/veterinary , Europe/epidemiology , Lumpy Skin Disease/epidemiology , Lumpy Skin Disease/pathology , Lumpy Skin Disease/transmission , Lumpy Skin Disease/virology , Lumpy skin disease virus/pathogenicity , Lumpy skin disease virus/ultrastructure , Skin/pathology , Skin/virology , Vasculitis/pathology , Vasculitis/veterinary , Vasculitis/virology
8.
Sci Rep ; 8(1): 16956, 2018 11 16.
Article in English | MEDLINE | ID: mdl-30446704

ABSTRACT

The re-emergence of poxviral zoonotic infections and the threat of bioterrorism emphasise the demand for effective antipoxvirus therapies. Here, we show that carbenoxolone, a pharmacological inhibitor of gap junction function and a compound widely used in cell culture, is capable of hindering the replication of Vaccinia virus, the prototypical poxvirus, in a gap junction-independent manner in a human keratinocyte cell line. Viral protein synthesis occurs in the presence of carbenoxolone but infectious virion formation is minimal, indicating that carbenoxolone blocks viral morphogenesis. Initial viability tests suggested that carbenoxolone was not toxic to cells. However, electron microscopic analysis of carbenoxolone treated cells revealed that it alters the cellular endomembrane system. This widespread ultrastructural damage prevents Vaccinia virus virion assembly. These results strengthen the need for thorough characterisation of the effects of antiviral compounds on the cellular ultrastructure.


Subject(s)
Carbenoxolone/pharmacology , Keratinocytes/drug effects , Virus Replication/drug effects , Anti-Ulcer Agents/pharmacology , Antiviral Agents/pharmacology , Cell Line , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cell Membrane/virology , Cell Survival/drug effects , Humans , Keratinocytes/cytology , Keratinocytes/virology , Microscopy, Electron, Transmission , Vaccinia virus/drug effects , Vaccinia virus/physiology , Virion/drug effects , Virion/metabolism
9.
J Gen Virol ; 99(5): 667-675, 2018 May.
Article in English | MEDLINE | ID: mdl-29595418

ABSTRACT

Cervical cancer is one of the leading causes of death in women worldwide and is etiologically linked to human papillomavirus (HPV) infection. Viral early proteins E6 and E7 manipulate cellular functions to promote the virus life cycle and are essential to the cellular transformation process. The innate immune system plays a pivotal role in the natural history of HPV infection. Among the various proteins that mediate the innate immune response, Toll-like receptors (TLRs) recognize pathogen-associated molecular patterns (PAMPs) and initiate the immune response. The objective of this study was to identify HPV E6 protein interaction partners in the TLR signalling pathway that may play a role in the immune response against HPV. Six TLR pathway proteins were shown to interact with HPV16 E6: myeloid differentiation primary response protein (MyD88), TIR domain-containing adapter molecule 1 (TRIF), interleukin-1 receptor-associated kinase-like (IRAK) 2, TNF receptor-associated factor (TRAF) 6, I-κB kinase beta (IKKß) and I-κB kinase epsilon (IKKε). The interaction site of IKKε with E6 is located in the region containing the enzyme catalytic site, suggesting an influence of E6 on the activation of IKKε target proteins. HPV16 E6 potentiated the activation of NF-κB by various TLR pathway members. These results suggest that HPV16 has the ability to interfere with components of the immune response, contributing to HPV carcinogenesis.

10.
Nat Commun ; 8: 14392, 2017 02 13.
Article in English | MEDLINE | ID: mdl-28194029

ABSTRACT

Many human cells can sense the presence of exogenous DNA during infection though the cytosolic DNA receptor cyclic GMP-AMP synthase (cGAS), which produces the second messenger cyclic GMP-AMP (cGAMP). Other putative DNA receptors have been described, but whether their functions are redundant, tissue-specific or integrated in the cGAS-cGAMP pathway is unclear. Here we show that interferon-γ inducible protein 16 (IFI16) cooperates with cGAS during DNA sensing in human keratinocytes, as both cGAS and IFI16 are required for the full activation of an innate immune response to exogenous DNA and DNA viruses. IFI16 is also required for the cGAMP-induced activation of STING, and interacts with STING to promote STING phosphorylation and translocation. We propose that the two DNA sensors IFI16 and cGAS cooperate to prevent the spurious activation of the type I interferon response.


Subject(s)
DNA/metabolism , Keratinocytes/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Phosphoproteins/metabolism , Cell Line , DNA Viruses/metabolism , Gene Expression , Humans , Immunity, Innate , Interferon-beta/genetics , Interferon-beta/metabolism , Keratinocytes/immunology , Mutation , Nuclear Proteins/genetics , Nucleotides, Cyclic/metabolism , Phosphoproteins/genetics , Phosphorylation , Protein Transport
11.
J Virol ; 90(22): 10120-10132, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27581988

ABSTRACT

Poxviruses, such as vaccinia virus (VACV), undertake a complex cytoplasmic replication cycle which involves morphogenesis through four distinct virion forms and includes a crucial wrapping step whereby intracellular mature virions (IMVs) are wrapped in two additional membranes to form intracellular enveloped virions (IEVs). To determine if cellular retrograde transport pathways are required for this wrapping step, we examined VACV morphogenesis in cells with reduced expression of the tetrameric tethering factor known as the GARP (Golgi-associated retrograde pathway), a central component of retrograde transport. VACV multistep replication was significantly impaired in cells transfected with small interfering RNA targeting the GARP complex and in cells with a mutated GARP complex. Detailed analysis revealed that depletion of the GARP complex resulted in a reduction in the number of IEVs, thereby linking retrograde transport with the wrapping of IMVs. In addition, foci of viral wrapping membrane proteins without an associated internal core accumulated in cells with a mutated GARP complex, suggesting that impaired retrograde transport uncouples nascent IMVs from the IEV membranes at the site of wrapping. Finally, small-molecule inhibitors of retrograde transport strongly suppressed VACV multistep growth in vitro and reduced weight loss and clinical signs in an in vivo murine model of systemic poxviral disease. This work links cellular retrograde transport pathways with the morphogenesis of poxviruses and identifies a panel of novel inhibitors of poxvirus replication. IMPORTANCE Cellular retrograde transport pathways traffic cargo from endosomes to the trans-Golgi network and are a key part of the intracellular membrane network. This work reveals that the prototypic poxvirus vaccinia virus (VACV) exploits cellular retrograde transport pathways to facilitate the wrapping of intracellular mature virions and therefore promote the production of extracellular virus. Inhibition of retrograde transport by small-molecule inhibitors reduced the replication of VACV in cell culture and alleviated disease in mice experimentally infected with VACV. This research provides fundamental new knowledge about the wrapping step of poxvirus morphogenesis, furthers our knowledge of the complex cellular retrograde pathways, and identifies a new group of antipoxvirus drugs.

12.
Virology ; 475: 66-73, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25462347

ABSTRACT

Vaccinia virus (VACV) is a large double-stranded DNA virus with a complex cytoplasmic replication cycle that exploits numerous cellular proteins. This work characterises the role of a proviral cellular protein, the small GTPase RAB1A, in VACV replication. Using siRNA, we identified RAB1A as required for the production of extracellular enveloped virions (EEVs), but not intracellular mature virions (IMVs). Immunofluorescence and electron microscopy further refined the role of RAB1A as facilitating the wrapping of IMVs to become intracellular enveloped virions (IEVs). This is consistent with the known function of RAB1A in maintenance of ER to Golgi transport. VACV can therefore be added to the growing list of viruses which require RAB1A for optimal replication, highlighting this protein as a broadly proviral host factor.


Subject(s)
Vaccinia virus/physiology , Virion/physiology , Virus Replication/physiology , rab1 GTP-Binding Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Humans , Vaccinia virus/genetics , Viral Plaque Assay , Virus Assembly , rab1 GTP-Binding Proteins/genetics
13.
PLoS One ; 9(6): e98431, 2014.
Article in English | MEDLINE | ID: mdl-24901222

ABSTRACT

Vaccinia virus (VACV) is a large, cytoplasmic, double-stranded DNA virus that requires complex interactions with host proteins in order to replicate. To explore these interactions a functional high throughput small interfering RNA (siRNA) screen targeting 6719 druggable cellular genes was undertaken to identify host factors (HF) influencing the replication and spread of an eGFP-tagged VACV. The experimental design incorporated a low multiplicity of infection, thereby enhancing detection of cellular proteins involved in cell-to-cell spread of VACV. The screen revealed 153 pro- and 149 anti-viral HFs that strongly influenced VACV replication. These HFs were investigated further by comparisons with transcriptional profiling data sets and HFs identified in RNAi screens of other viruses. In addition, functional and pathway analysis of the entire screen was carried out to highlight cellular mechanisms involved in VACV replication. This revealed, as anticipated, that many pro-viral HFs are involved in translation of mRNA and, unexpectedly, suggested that a range of proteins involved in cellular transcriptional processes and several DNA repair pathways possess anti-viral activity. Multiple components of the AMPK complex were found to act as pro-viral HFs, while several septins, a group of highly conserved GTP binding proteins with a role in sequestering intracellular bacteria, were identified as strong anti-viral VACV HFs. This screen has identified novel and previously unexplored roles for cellular factors in poxvirus replication. This advancement in our understanding of the VACV life cycle provides a reliable knowledge base for the improvement of poxvirus-based vaccine vectors and development of anti-viral theraputics.


Subject(s)
Host-Pathogen Interactions/genetics , RNA Interference , RNA, Small Interfering/genetics , Vaccinia virus/physiology , Vaccinia/genetics , Vaccinia/virology , Virus Replication , Gene Expression Regulation , Gene Knockdown Techniques , High-Throughput Screening Assays , Humans , Reproducibility of Results , Signal Transduction , Transcription, Genetic , Vaccinia/metabolism
14.
J Virol ; 88(7): 3664-77, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24429366

ABSTRACT

UNLABELLED: Tumor necrosis factor receptor (TNFR)-associated factor 2 (TRAF2) is a pivotal intracellular mediator of signaling pathways downstream of TNFR1 and -2 with known pro- and antiviral effects. We investigated its role in the replication of the prototype poxvirus vaccinia virus (VACV). Loss of TRAF2 expression, either through small interfering RNA treatment of HeLa cells or through genetic knockout in murine embryonic fibroblasts (MEFs), led to significant reductions in VACV growth following low-multiplicity infection. In single-cycle infections, there was delayed production of both early and late VACV proteins as well as accelerated virus-induced alterations to cell morphology, indicating that TRAF2 influences early stages of virus replication. Consistent with an early role, uncoating assays showed normal virus attachment but delayed virus entry in the absence of TRAF2. Although alterations to c-Jun N-terminal kinase (JNK) signaling were apparent in VACV-infected TRAF2(-/-) MEFs, treatment of wild-type cells with a JNK inhibitor did not affect virus entry. Instead, treatment with an inhibitor of endosomal acidification greatly reduced virus entry into TRAF2(-/-) MEFs, suggesting that VACV is reliant on the endosomal route of entry in the absence of TRAF2. Thus, TRAF2 is a proviral factor for VACV that plays a role in promoting efficient viral entry, most likely via the plasma membrane. IMPORTANCE: Tumor necrosis factor receptor-associated factors (TRAFs) are key facilitators of intracellular signaling with roles in innate and adaptive immunity and stress responses. We have discovered that TRAF2 is a proviral factor in vaccinia virus replication in both HeLa cells and mouse embryonic fibroblasts and that its influence is exercised through promotion of efficient virus entry.


Subject(s)
Host-Pathogen Interactions , TNF Receptor-Associated Factor 2/metabolism , Vaccinia virus/physiology , Virus Internalization , Animals , Cell Line , Epithelial Cells/virology , Fibroblasts/virology , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Mice , TNF Receptor-Associated Factor 2/genetics
15.
J Biol Chem ; 282(34): 24759-66, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17573354

ABSTRACT

Innate immune responses are critical in controlling viral infections. Viral proteins and nucleic acids have been shown to be recognized by pattern recognition receptors of the Toll-like receptor (TLR) family, triggering downstream signaling cascades that lead to cellular activation and cytokine production. Viral DNA is sensed by TLR9, and TLRs 3, 7, and 8 have been implicated in innate responses to RNA viruses by virtue of their ability to sense double-stranded (ds) RNA (TLR3) or single-stranded RNA (murine TLR7 and human TLR8). Viral and synthetic dsRNAs have also been shown to be a potent adjuvant, promoting enhanced adaptive immune responses, and this property is also dependent on their recognition by TLR3. It has recently been shown that mRNA that is largely single-stranded is a ligand for TLR3. Here we have investigated the ability of single-stranded homopolymeric nucleic acids to induce innate responses by murine immune cells. We show for the first time that polyinosinic acid (poly(I)) activates B lymphocytes, dendritic cells, and macrophages and that these responses are dependent on the expression of both TLR3 and the adaptor molecule, Toll/IL-1 receptor domain-containing adaptor inducing IFN-beta (TRIF). We therefore conclude that TLR3 is able to sense both single-stranded RNA and dsRNA.


Subject(s)
Poly I/metabolism , RNA, Double-Stranded/chemistry , RNA/chemistry , Toll-Like Receptor 3/metabolism , Animals , B-Lymphocytes/metabolism , Bronchi/cytology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Epithelial Cells/metabolism , Humans , Ligands , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
16.
Mol Immunol ; 42(8): 859-67, 2005 May.
Article in English | MEDLINE | ID: mdl-15829275

ABSTRACT

The discovery of Toll-like receptors (TLR) has revolutionised our understanding of innate immunity. Numerous reviews have been written on the subject in the past few years. Here, we review the evidence that TLRs are involved in sensing and initiating anti-viral responses. There are now three strong lines of evidence that support such a role for TLRs. Firstly, TLRs 'recognise' virally derived molecules and are required for various virus-induced cellular effects. Secondly, TLRs trigger anti-viral signalling pathways leading to the induction of the interferon response. Thirdly, viral immune strategies employed against TLRs have been identified.


Subject(s)
Membrane Glycoproteins/immunology , Receptors, Cell Surface/immunology , Virus Diseases/immunology , Viruses/immunology , Animals , DNA-Binding Proteins/metabolism , Humans , Interferon Regulatory Factor-3 , Membrane Glycoproteins/metabolism , NF-kappa B/immunology , NF-kappa B/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/immunology , Signal Transduction/physiology , Toll-Like Receptors , Transcription Factors/metabolism , Virus Diseases/metabolism
17.
J Exp Med ; 201(6): 1007-18, 2005 Mar 21.
Article in English | MEDLINE | ID: mdl-15767367

ABSTRACT

Viral immune evasion strategies target key aspects of the host antiviral response. Recently, it has been recognized that Toll-like receptors (TLRs) have a role in innate defense against viruses. Here, we define the function of the vaccinia virus (VV) protein A46R and show it inhibits intracellular signalling by a range of TLRs. TLR signalling is triggered by homotypic interactions between the Toll-like-interleukin-1 resistance (TIR) domains of the receptors and adaptor molecules. A46R contains a TIR domain and is the only viral TIR domain-containing protein identified to date. We demonstrate that A46R targets the host TIR adaptors myeloid differentiation factor 88 (MyD88), MyD88 adaptor-like, TIR domain-containing adaptor inducing IFN-beta (TRIF), and the TRIF-related adaptor molecule and thereby interferes with downstream activation of mitogen-activated protein kinases and nuclear factor kappaB. TRIF mediates activation of interferon (IFN) regulatory factor 3 (IRF3) and induction of IFN-beta by TLR3 and TLR4 and suppresses VV replication in macrophages. Here, A46R disrupted TRIF-induced IRF3 activation and induction of the TRIF-dependent gene regulated on activation, normal T cell expressed and secreted. Furthermore, we show that A46R is functionally distinct from another described VV TLR inhibitor, A52R. Importantly, VV lacking the A46R gene was attenuated in a murine intranasal model, demonstrating the importance of A46R for VV virulence.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Antigens, Differentiation/metabolism , MAP Kinase Signaling System/physiology , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Vaccinia virus/metabolism , Viral Proteins/metabolism , ATP-Binding Cassette Transporters , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Cell Line , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation, Viral/genetics , Gene Expression Regulation, Viral/physiology , Humans , Interferon Regulatory Factor-3 , Interferon-beta/biosynthesis , Lymphocyte Activation/genetics , Lymphocyte Activation/physiology , MAP Kinase Signaling System/genetics , Mice , Molecular Sequence Data , Myeloid Differentiation Factor 88 , Periplasmic Binding Proteins , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , T-Lymphocytes/physiology , Toll-Like Receptor 3 , Toll-Like Receptor 4 , Toll-Like Receptors , Transcription Factors/metabolism , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Viral Proteins/genetics , Virus Diseases/genetics , Virus Diseases/physiopathology , Virus Replication/genetics , Virus Replication/physiology
18.
J Exp Med ; 197(3): 343-51, 2003 Feb 03.
Article in English | MEDLINE | ID: mdl-12566418

ABSTRACT

Toll-like receptors (TLRs) are crucial in the innate immune response to pathogens, in that they recognize and respond to pathogen associated molecular patterns, which leads to activation of intracellular signaling pathways and altered gene expression. Vaccinia virus (VV), the poxvirus used to vaccinate against smallpox, encodes proteins that antagonize important components of host antiviral defense. Here we show that the VV protein A52R blocks the activation of the transcription factor nuclear factor kappa B (NF-kappa B) by multiple TLRs, including TLR3, a recently identified receptor for viral RNA. A52R associates with both interleukin 1 receptor-associated kinase 2 (IRAK2) and tumor necrosis factor receptor-associated factor 6 (TRAF6), two key proteins important in TLR signal transduction. Further, A52R could disrupt signaling complexes containing these proteins. A virus deletion mutant lacking the A52R gene was attenuated compared with wild-type and revertant controls in a murine intranasal model of infection. This study reveals a novel mechanism used by VV to suppress the host immunity. We demonstrate viral disabling of TLRs, providing further evidence for an important role for this family of receptors in the antiviral response.


Subject(s)
Adaptor Proteins, Signal Transducing , Drosophila Proteins , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/immunology , Receptors, Cell Surface/immunology , Vaccinia virus/immunology , Vaccinia virus/pathogenicity , Viral Proteins/immunology , Animals , Carrier Proteins/metabolism , Cell Line , Female , Gene Deletion , Genes, Viral , Humans , Immune Tolerance , Interleukin-1 Receptor-Associated Kinases , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Protein Kinases/metabolism , Proteins/metabolism , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction , TNF Receptor-Associated Factor 6 , Toll-Like Receptor 3 , Toll-Like Receptors , Vaccinia virus/genetics , Viral Proteins/genetics , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...