Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Colin J. McInnes is active.

Publication


Featured researches published by Colin J. McInnes.


Journal of Virology | 2000

Orf Virus Encodes a Novel Secreted Protein Inhibitor of Granulocyte-Macrophage Colony-Stimulating Factor and Interleukin-2

David Deane; Colin J. McInnes; Ann Percival; Ann R. Wood; Jackie Thomson; Andrea Lear; Janice Gilray; Stephen B. Fleming; Andrew A. Mercer; David M. Haig

ABSTRACT The parapoxvirus orf virus encodes a novel soluble protein inhibitor of ovine granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-2 (IL-2). The GM-CSF- and IL-2-inhibitory factor (GIF) gene was expressed as an intermediate-late viral gene in orf virus-infected cells. GIF formed homodimers and tetramers in solution, and it bound ovine GM-CSF with a Kd of 369 pM and ovine IL-2 with a Kd of 1.04 nM. GIF did not bind human GM-CSF or IL-2 in spite of the fact that orf virus is a human pathogen. GIF was detected in afferent lymph plasma draining the skin site of orf virus reinfection and was associated with reduced levels of lymph GM-CSF. GIF expression by orf virus indicates that GM-CSF and IL-2 are important in host antiviral immunity.


Expert Review of Anti-infective Therapy | 2009

Orf: an update on current research and future perspectives

Madhusudan Hosamani; Alessandra Scagliarini; Veerakyathappa Bhanuprakash; Colin J. McInnes; Raj Kumar Singh

Orf is one of the most widespread viral diseases worldwide, affecting mostly small ruminants and, sometimes, other species, including wild animals. Of late, there have been an increasing number of reports of new species being affected by the disease, implying a dynamic host–pathogen interaction. The causative agent, orf virus, has been extensively investigated over recent years, owing to its zoonotic importance and ability to cross-infect other species sporadically. The evasive mechanisms that the virus has developed to adapt and grow in the presence of an active immune response helps to explain the ability of the virus to repeatedly reinfect the same host. The apparent diversity in the antigenic/immune targets of different orf virus strains involved in such repeat infections may also be contributing factors. Exposure of animals to stress or immunosupression as a result of therapy or primary viral infection can accentuate the severity of disease. Genes homologous to host cytokines or their antagonists, and which contribute to viral virulence, have been found in the viral genome. A combination of electron microscopy, histology and PCR is the most accurate laboratory approach for confirmation of the disease, although clinical signs are often typical. However, some infections may be confounded by similar clinical manifestations caused by other infections. This review presents, in brief, a recent understanding of the virus at the host–pathogen level, molecular biology of the virus, disease epidemiology, clinical manifestations in man and animals, diagnostic procedures, and the economic and environmental impact of the disease.


Virus Research | 2002

Immunity and counter-immunity during infection with the parapoxvirus orf virus

David M. Haig; Colin J. McInnes

Orf virus is a DNA parapoxvirus that causes orf, an acute debilitating skin disease of sheep, goats and humans. In sheep, a vigorous immune response involving neutrophils, dermal dendritic cells, T cells, B cells and antibody is generated after infection. CD4(+) T cells, IFN-gamma and to a lesser extent CD8(+) T cells are involved in partial protection against infection. In spite of this, orf virus can repeatedly infect sheep albeit with reduced lesion size and time to resolution compared to primary infection. This is due at least in part to the action of virus immuno-modulator proteins that interfere with host immune and inflammatory responses. These include: an interferon resistance protein; a viral orthologue of mammalian IL-10 (vIL-10) that is an anti-inflammatory cytokine; and a novel inhibitor of the cytokines GM-CSF and IL-2 (GIF). The virus also encodes a virulence protein that is an orthologue of mammalian vascular endothelial growth factor. The study of the immuno-modulator proteins provides an insight into disease pathogenesis and important elements of a host protective response. This information will be used to devise a rational disease control strategy.


Veterinary Immunology and Immunopathology | 1998

Early immunopathological events in experimental ovine paratuberculosis.

I Begara-McGorum; L.A Wildblood; C.J Clarke; K.M Connor; K Stevenson; Colin J. McInnes; J.M Sharp; D.G Jones

An experimental oral infection of neonatal (< 2 weeks old) lambs with a cervine isolate of Mycobacterium avium subspecies paratuberculosis (M.a. paratuberculosis), the causal agent of ruminant paratuberculosis (Johnes disease) was used to investigate bacteriological, histopathological and immunological changes during the early (up to 8 weeks) post-infection phase. In vitro culture for mycobacteria was positive in one faecal and three mesenteric lymph node (MLN) samples from the eight infected lambs. All mycobacterial isolates from MLN were identified as M.a. paratuberculosis by polymerase chain reaction (PCR). Small-to-medium sized focal granulomata were observed in jejunal (JPP) and ileal Peyers patches (IPP) from four of the eight infected lambs. Compared with controls, JPP from all infected lambs had significantly (p < 0.05) higher proportions of CD8+ and CD2+ lymphocytes, and there were significantly (p < 0.05) fewer cells expressing B lymphocyte-associated markers in IPP and MLN. The T/B cell ratio was significantly (p < 0.05) increased in both JPP and MLN from infected lambs. The expression of a range of genes for cytokines was examined using specific reverse transcriptase PCR (RT-PCR) amplification of messenger RNA (mRNA) template isolated from MLN, JPP and IPP from both groups of animals. Densitometric analyses indicated that, in infected animals, MLN expressed significantly (p < 0.05) more mRNA for TNF-alpha: JPP had significantly increased (p < 0.05) mRNA for GM-CSF and significantly decreased (p < 0.05) mRNA for IL-4 and IFN-gamma. Infected lambs had significantly (p < 0.05) decreased titres of both circulating IgG and gut mycobacteria-associated IgG antibody. Infection was not associated with any consistent changes in lymphocyte reactivity to specific mycobacterial antigens, IFN-gamma release into supernatants from in vitro intestinal lymphocyte cultures or gut IgA antibody levels.


Ecohealth | 2008

Poxviral disease in red squirrels Sciurus vulgaris in the UK: spatial and temporal trends of an emerging threat.

Anthony W. Sainsbury; Robert Deaville; Becki Lawson; W. A. Cooley; Stephan S. J. Farelly; M.J. Stack; Paul Duff; Colin J. McInnes; John Gurnell; P.H. Russell; Stephen Rushton; Dirk U. Pfeiffer; Peter Nettleton; Peter W. W. Lurz

The squirrel poxvirus (SQPV) is the probable mediator of apparent competition between the introduced invading gray squirrel (Sciurus carolinensis) and the red squirrel (Sciurus vulgaris) in the UK, and modeling studies have shown that this viral disease has had a significant impact on the decline of the red squirrel in the UK. However, given our limited understanding of the epidemiology of the disease, and more generally the effects of invasive species on parasite ecology, there is a need to investigate the transmission dynamics and the relative pathogenicity of the virus between species. We aimed to increase our knowledge of these processes through an empirical study in which we: (i) used pathological signs and transmission electron microscopy (TEM) to diagnose SQPV disease in red squirrels found dead during scanning surveillance between 1993 and 2005; (ii) detected antibody to SQPV using an enzyme-linked immunosorbent assay (ELISA) in the same animals; and (iii) mapped cases of the disease, and the gray squirrel distribution, using a geographical information system. We analyzed the distribution of cases of SQPV disease according to woodland type, a measure of squirrel density. SQPV disease occurred only in areas of England also inhabited by seropositive gray squirrels, and as the geographical range of gray squirrels expanded, SQPV disease occurred in these new gray squirrel habitats, supporting a role for the gray squirrel as a reservoir host of the virus. There was a delay between the establishment of invading gray squirrels and cases of the disease in red squirrels which implies gray squirrels must reach a threshold number or density before the virus is transmitted to red squirrels. The spatial and temporal trend in SQPV disease outbreaks suggested that SQPV disease will have a significant effect on Scottish populations of red squirrels within 25 years. The even spread of cases of disease across months suggested a direct rather than vector-borne transmission route is more likely. Eight juvenile and sub-adult free-living red squirrels apparently survived exposure to SQPV by mounting an immune response, the first evidence of immunity to SQPV in free-living red squirrels, which possibly suggests a changing host-parasite relationship and that the use of a vaccine may be an effective management tool to protect remnant red squirrel populations.


Immunology | 2001

The orf virus OV20.0L gene product is involved in interferon resistance and inhibits an interferon-inducible, double-stranded RNA-dependent kinase.

David M. Haig; Colin J. McInnes; Jackie Thomson; Ann R. Wood; K Bunyan; Andrew A. Mercer

The parapoxvirus orf virus was resistant to type 1 (IFN‐α) and type 2 (IFN‐γ) interferons in cultures of ovine cells. The recently identified orf virus OV20.0L gene exhibits 31% predicted amino acid identity to the vaccinia virus E3L interferon‐resistance gene, and is referred to as the (putative) orf virus interferon‐resistance gene (OVIFNR). The objective of this study was to determine whether OVIFNR was involved in interferon resistance. Recombinant OVIFNR as a thioredoxin fusion protein (OVIFNR–Tx) inhibited the activation (by autophosphorylation) of an interferon‐inducible, double‐stranded (ds) RNA‐dependent kinase (PKR) of sheep, which was shown to bind dsRNA (poly I:C). PKR in other species is involved in the inhibition of protein synthesis as part of the antiviral state in infected cells. Virus‐infected cell lysates, but not control lysates, from cells grown in the presence of cytosine arabinoside also contained PKR inhibitory activity, which indicated that the inhibitory activity was associated with early viral gene expression. Significantly, the OVIFNR gene expressed in interferon‐treated ovine fibroblasts protected the unrelated Semliki Forest virus from the antiviral effect of both type 1 and type 2 interferons. Taken together, the results indicate that the OVIFNR gene functions as an interferon‐resistance gene, the product of which inhibits PKR in a similar way to the vaccinia virus E3L gene product.


Virus Genes | 1998

Orf Virus Encodes a Homolog of the Vaccinia Virus Interferon-Resistance Gene E3L

Colin J. McInnes; Ann R. Wood; Andrew A. Mercer

A homolog of the vaccinia virus (VAC) interferon resistance gene E3L has been discovered in orf virus strain NZ-2, a parapoxvirus that infects sheep, goats and humans. The gene is located 20 kb from the left terminus of the orf virus genome and is transcribed towards this terminus. RNase protection studies have been used to define the limits of the gene and Northern analysis revealed that it is expressed early in infection. The predicted amino acid sequence of the orf virus protein shares 31% identity (57% similarity) with the VAC E3L protein. Four of the six residues identified as being essential to dsRNA binding in the vaccinia virus protein are conserved in the orf virus protein whilst the other two amino acid changes are conservative substitutions. The orf virus gene has been sequenced in two other orf virus strains which vary markedly in their ability to produce experimental lesions in vivo. Their predicted protein sequences vary by less than 3% from t he NZ-2 protein. The recombinant orf virus protein, expressed as a fusion protein in E. coli, bound double-stranded (ds)RNA but not dsDNA, single-stranded (ss)DNA or ssRNA . This is the first demonstration of a VAC E3L-like gene encoded by a parapoxvirus.


Gene | 1991

Cloning and expression of a cDNA encoding ovine granulocyte-macrophage colony-stimulating factor.

Colin J. McInnes; David M. Haig

A cDNA encoding ovine granulocyte-macrophage colony-stimulating factor (GM-CSF) has been cloned using the polymerase chain reaction. The nucleotide sequence is approx. 93% identical to the published bovine GM-CSF-encoding sequence, 84% to the human sequence and 73% to the murine sequence. The deduced amino acid sequence of the ovine GM-CSF protein was found to be 80% identical to both the human and bovine proteins and 57% to the murine protein. Transient expression of recombinant ovine GM-CSF in COS-1 cells was obtained and its biological activity investigated in a bone-marrow colony-forming assay. Ovine GM-CSF was found to promote the formation of granulocyte-macrophage colonies as well as eosinophil, neutrophil and monocyte/macrophage colonies, an activity characteristic of GM-CSF in other species. Recombinant human GM-CSF was found to have no proliferative effect on ovine bone-marrow cells.


Journal of General Virology | 2010

The genome of pseudocowpoxvirus: comparison of a reindeer isolate and a reference strain.

Maria Hautaniemi; Norihito Ueda; Jarno Tuimala; Andrew A. Mercer; Juhani Lahdenperä; Colin J. McInnes

Parapoxviruses (PPV), of the family Poxviridae, cause a pustular cutaneous disease in sheep and goats (orf virus, ORFV) and cattle (pseudocowpoxvirus, PCPV and bovine papular stomatitis virus, BPSV). Here, we present the first genomic sequence of a reference strain of PCPV (VR634) along with the genomic sequence of a PPV (F00.120R) isolated in Finland from reindeer (Rangifer tarandus tarandus). The F00.120R and VR634 genomes are 135 and 145 kb in length and contain 131 and 134 putative genes, respectively, with their genome organization being similar to that of other PPVs. The predicted proteins of F00.120R and VR634 have an average amino acid sequence identity of over 95%, whereas they share only 88 and 73% amino acid identity with the ORFV and BPSV proteomes, respectively. The most notable differences were found near the genome termini. F00.120R lacks six and VR634 lacks three genes seen near the right terminus of other PPVs. Four genes at the left end of F00.120R and one in the middle of both genomes appear to be fragmented paralogues of other genes within the genome. VR634 has larger than expected inverted terminal repeats possibly as a result of genomic rearrangements. The high G+C content (64%) of these two viruses along with amino acid sequence comparisons and whole genome phylogenetic analyses confirm the classification of PCPV as a separate species within the genus Parapoxvirus and verify that the virus responsible for an outbreak of contagious stomatitis in reindeer over the winter of 1999-2000 can be classified as PCPV.


Journal of General Virology | 2002

Vascular endothelial growth factors encoded by Orf virus show surprising sequence variation but have a conserved, functionally relevant structure

Andrew A. Mercer; Lyn M. Wise; Alessandra Scagliarini; Colin J. McInnes; Mathias Büttner; Hanns-Joachim Rziha; Catherine A. McCaughan; Stephen B. Fleming; Norihito Ueda; Peter Nettleton

The first report of a vascular endothelial growth factor (VEGF)-like gene in Orf virus included the surprising observation that the genes from two isolates (NZ2 and NZ7) shared only 41.1% amino acid sequence identity. We have examined this sequence disparity by determining the VEGF gene sequence of 21 isolates of Orf virus derived from diverse sources. Most isolates carried NZ2-like VEGF genes but their predicted amino acid sequences varied by up to 30.8% with an average amino acid identity between pairs of NZ2-like sequences of 86.1%. This high rate of sequence variation is more similar to interspecies than intraspecies variability. In contrast, only three isolates carried an NZ7-like VEGF gene and these varied from the NZ7 sequence by no more than a single nucleotide. The VEGF family are ligands for a set of tyrosine kinase receptors. The viral VEGFs are unique among the family in that they recognize VEGF receptor 2 (VEGFR-2) but not VEGFR-1 or VEGFR-3. Comparisons of the viral VEGFs with other family members revealed some correlations between conserved residues and the ability to recognize specific VEGF receptors. Despite the sequence variations, structural predictions for the viral VEGFs were very similar to each other and to the structure determined by X-ray crystallography for human VEGF-A. Structural modelling also revealed that a groove seen in the VEGF-A homodimer and believed to play a role in its binding to VEGFR-1 is blocked in the viral VEGFs. This may contribute to the inability of the viral VEGFs to bind VEGFR-1.

Collaboration


Dive into the Colin J. McInnes's collaboration.

Top Co-Authors

Avatar

David M. Haig

University of Nottingham

View shared research outputs
Top Co-Authors

Avatar

David J. Everest

Veterinary Laboratories Agency

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jayne Hope

University of Edinburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthony W. Sainsbury

Zoological Society of London

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge