Andrew Skelton
Newcastle University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Andrew Skelton.
BMC Medical Genetics | 2015
Colin Shepherd; Andrew Skelton; Michael D. Rushton; Louise N. Reynard; John Loughlin
BackgroundOsteoarthritis (OA) is a painful, debilitating disease characterised by loss of articular cartilage with concurrent changes in other tissues of the synovial joint. Genetic association studies have shown that a number of common variants increase the risk of developing OA. Investigating their activity can uncover novel causal pathways and potentially highlight new treatment targets. One of the reported OA association signals is marked by the single nucleotide polymorphism (SNP) rs11842874 at chromosome 13q34. rs11842874 is positioned within a small linkage disequilibrium (LD) block within intron 4 of MCF2L, a gene encoding guanine-nucleotide exchange factor DBS. There are no non-synonymous SNPs that correlate with this association signal and we therefore set out to assess whether its effect on OA susceptibility is mediated by alteration of MCF2L expression.MethodsNucleic acid was extracted from cartilage, synovial membrane or infrapatellar fat pad tissues from OA patients. Expression of MCF2L was measured by quantitative PCR and RNA-sequencing whilst the presence of DBS was studied using immunohistochemistry. The functional effect of SNPs within the 13q34 locus was assessed using public databases and in vitro using luciferase reporter analysis.ResultsMCF2L gene and protein expression are detectable in joint tissues, with quantitative differences in the expression of the gene and in the transcript isoforms expressed between the tissues tested. There is an expression quantitative trait locus (eQTL) operating within synovial membrane tissue, with possession of the risk-conferring A allele of rs11842874 correlating with increased MCF2L expression. SNPs within the rs11842874 LD block reside within transcriptional regulatory elements and their direct analysis reveals that several show quantitative differences in regulatory activity at the allelic level.ConclusionsMCF2L is subject to a cis-acting eQTL in synovial membrane that correlates with the OA association signal. This signal contains several functional SNPs that could account for the susceptibility and which therefore merit further investigation. As far as we are aware, this is the first example of an OA susceptibility locus operating as an eQTL in synovial membrane tissue but not in cartilage.
Journal of Biological Chemistry | 2017
Chun Ming Chan; Christopher D. Macdonald; Gary J. Litherland; David J. Wilkinson; Andrew Skelton; G. Nicholas Europe-Finner; Andrew D. Rowan
Irreversible breakdown of cartilage extracellular matrix (ECM) by the collagenase matrix metalloproteinase 13 (MMP13) represents a key event in osteoarthritis (OA) progression. Although inflammation is most commonly associated with inflammatory joint diseases, it also occurs in OA and is thus relevant to the prevalent tissue destruction. Here, inflammation generates a cFOS AP-1 early response that indirectly affects MMP13 gene expression. To ascertain a more direct effect on prolonged MMP13 production we examined the potential molecular events occurring between the rapid, transient expression of cFOS and the subsequent MMP13 induction. Importantly, we show MMP13 mRNA expression is mirrored by nascent hnRNA transcription. Employing ChIP assays, cFOS recruitment to the MMP13 promoter occurs at an early stage prior to gene transcription and that recruitment of transcriptional initiation markers also correlated with MMP13 expression. Moreover, protein synthesis inhibition following early FOS expression resulted in a significant decrease in MMP13 expression thus indicating a role for different regulatory factors modulating expression of the gene. Subsequent mRNA transcriptome analyses highlighted several genes induced soon after FOS that could contribute to MMP13 expression. Specific small interfering RNA-mediated silencing highlighted that ATF3 was as highly selective for MMP13 as cFOS. Moreover, ATF3 expression was AP-1(cFOS/cJUN)-dependent and expression levels were maintained after the early transient cFOS response. Furthermore, ATF3 bound the proximal MMP13 AP-1 motif in stimulated chondrocytes at time points that no longer supported binding of FOS. Consequently, these findings support roles for both cFOS (indirect) and ATF3 (direct) in effecting MMP13 transcription in human chondrocytes.
PLOS ONE | 2016
Rachel C. Williams; Andrew Skelton; Stephen Todryk; Andrew D. Rowan; Philip M. Preshaw; John J. Taylor
Introduction Gingival fibroblast-mediated extracellular matrix remodelling is implicated in the pathogenesis of periodontitis, yet the stimuli that regulate this response are not fully understood. The immunoregulatory adipokine leptin is detectable in the gingiva, human gingival fibroblasts express functional leptin receptor mRNA and leptin is known to regulate extracellular matrix remodelling responses in cardiac fibroblasts. We therefore hypothesised that leptin would enhance matrix metalloproteinase secretion in human gingival fibroblasts. Methods and Results We used in vitro cell culture to investigate leptin signalling and the effect of leptin on mRNA and protein expression in human gingival fibroblasts. We confirmed human gingival fibroblasts expressed cell surface leptin receptor, found leptin increased matrix metalloproteinase-1, -3, -8 and -14 expression in human gingival fibroblasts compared to unstimulated cells, and observed that leptin stimulation activated MAPK, STAT1/3 and Akt signalling in human gingival fibroblasts. Furthermore, leptin synergised with IL-1 or the TLR2 agonist pam2CSK4 to markedly enhance matrix metalloproteinase-1 and -3 production by human gingival fibroblasts. Signalling pathway inhibition demonstrated ERK was required for leptin-stimulated matrix metalloproteinase-1 expression in human gingival fibroblasts; whilst ERK, JNK, p38 and STAT3 were required for leptin+IL-1- and leptin+pam2CSK4-induced matrix metalloproteinase-1 expression. A genome-wide expression array and gene ontology analysis confirmed genes differentially expressed in leptin+IL-1-stimulated human gingival fibroblasts (compared to unstimulated cells) were enriched for extracellular matrix organisation and disassembly, and revealed that matrix metalloproteinase-8 and -12 were also synergistically upregulated by leptin+IL-1 in human gingival fibroblasts. Conclusions We conclude that leptin selectively enhances the expression and secretion of certain matrix metalloproteinases in human gingival fibroblasts, and suggest that gingival fibroblasts may have an ECM-degrading phenotype during conditions of hyperleptinaemia (e.g., obesity, type 2 diabetes mellitus, exogenous leptin therapy).
Arthritis & Rheumatism | 2018
Colin Shepherd; Dongxing Zhu; Andrew Skelton; Jennifer Combe; Harrison Threadgold; Linyi Zhu; Tonia L. Vincent; Paul Stuart; Louise N. Reynard; John Loughlin
To identify the functional single‐nucleotide polymorphisms (SNPs) and mechanisms conferring increased risk of hand osteoarthritis (OA) at the ALDH1A2 locus, which is a retinoic acid regulatory gene.
Connective Tissue Research | 2017
Ingrid Meulenbelt; Nidhi Bhutani; W. den Hollander; U. Oppermann; Louise N. Reynard; Andrew Skelton; David Young; F Beier; John Loughlin
ABSTRACT Osteoarthritis (OA) is a major clinical problem across the world, in part due to the lack of disease-modifying drugs resulting, to a significant degree, from our incomplete understanding of the underlying molecular mechanisms of the disease. Emerging evidence points to a role of epigenetics in the pathogenesis of OA, but research in this area is still in its early stages. In order to summarize current knowledge and to facilitate the potential coordination of future research activities, the first international workshop on the epigenetics of OA was held in Amsterdam in October 2015. Recent findings on DNA methylation and hydroxymethylation, histone modifications, noncoding RNAs, and other epigenetic mechanisms were presented and discussed. The workshop demonstrated the advantage of bringing together those working in this nascent field and highlights from the event are summarized in this report in the form of summaries from invited speakers and organizers.
Arthritis & Rheumatism | 2018
Nishanthi Thalayasingam; Nisha Nair; Andrew Skelton; Jonathan Massey; Amy E. Anderson; Alex Clark; Julie Diboll; Dennis Lendrem; Louise N. Reynard; Heather J. Cordell; Stephen Eyre; John D. Isaacs; Anne Barton; Arthur G. Pratt
Rheumatoid arthritis (RA) is a genetically complex disease of immune dysregulation. This study sought to gain further insight into the genetic risk mechanisms of RA by conducting an expression quantitative trait locus (eQTL) analysis of confirmed genetic risk loci in CD4+ T cells and B cells from carefully phenotyped patients with early arthritis who were naive to therapeutic immunomodulation.
Frontiers in Immunology | 2018
Faye A. H. Cooles; Amy E. Anderson; Andrew Skelton; Arthur G. Pratt; Mariola Kurowska-Stolarska; Iain B. McInnes; Catharien M. U. Hilkens; John D. Isaacs
Objective Dendritic cells (DCs) are key orchestrators of immune function. To date, rheumatoid arthritis (RA) researchers have predominantly focused on a potential pathogenic role for CD1c+ DCs. In contrast, CD141+ DCs and plasmacytoid DCs (pDCs) have not been systematically examined, at least in early RA. In established RA, the role of pDCs is ambiguous and, since disease duration and treatment both impact RA pathophysiology, we examined pDCs, and CD1c+ and CD141+ conventional DCs (cDCs), in early, drug-naïve RA (eRA) patients. Methods We analyzed the frequency and phenotype of pDCs, CD1c+, and CD141+ DCs from eRA patients and compared findings with healthy controls. In parallel, we performed transcriptional analysis of >600 immunology-related genes (Nanostring) from peripheral blood pDCs, CD1c+ DCs, B cells, T cells, and monocytes. Results All DC subsets were reduced in eRA (n = 44) compared with healthy controls (n = 30) and, for pDCs, this was most marked in seropositive patients. CD141+ and CD1c+ DCs, but not pDCs, had a comparatively activated phenotype at baseline (increased CD86) and CD1c+ DC frequency inversely associated with disease activity. All DC frequencies remained static 12 months after initiation of immunomodulatory therapy despite a fall in activation markers (e.g., HLA-DR, CD40). There was no association between the whole blood interferon gene signature (IGS) and pDC or CD1c+ DC parameters but an inverse association between CD141+ DC frequency and IGS was noted. Furthermore, IFN-I and IFN-III mRNA transcripts were comparable between eRA pDC and other leukocyte subsets (B cells, CD4+, and CD8+ T cells and monocytes) with no obvious circulating cellular source of IFN-I or IFN-III. Transcriptomic analysis suggested increased pDC and CD1c+ DC proliferation in eRA; pDC differentially expressed genes also suggested enhanced tolerogenic function, whereas for CD1c+ DCs, pro-inflammatory transcripts were upregulated. Discussion This is the first detailed examination of DC subsets in eRA peripheral blood. Compared with CD1c+ DCs, pDCs are less activated and may be skewed toward tolerogenic functions. CD141+ DCs may be implicated in RA pathophysiology. Our findings justify further investigation of early RA DC biology.
Development | 2017
M.J. Barter; Rodolfo Gómez; Sam Hyatt; Kat Cheung; Andrew Skelton; Yaobo Xu; Ian M. Clark; David Young
Long non-coding RNAs (lncRNAs) are expressed in a highly tissue-specific manner and function in various aspects of cell biology, often as key regulators of gene expression. In this study, we established a role for lncRNAs in chondrocyte differentiation. Using RNA sequencing we identified a human articular chondrocyte repertoire of lncRNAs from normal hip cartilage donated by neck of femur fracture patients. Of particular interest are lncRNAs upstream of the master chondrocyte transcription factor SOX9 locus. SOX9 is an HMG-box transcription factor that plays an essential role in chondrocyte development by directing the expression of chondrocyte-specific genes. Two of these lncRNAs are upregulated during chondrogenic differentiation of mesenchymal stem cells (MSCs). Depletion of one of these lncRNAs, LOC102723505, which we termed ROCR (regulator of chondrogenesis RNA), by RNA interference disrupted MSC chondrogenesis, concomitant with reduced cartilage-specific gene expression and incomplete matrix component production, indicating an important role in chondrocyte biology. Specifically, SOX9 induction was significantly ablated in the absence of ROCR, and overexpression of SOX9 rescued the differentiation of MSCs into chondrocytes. Our work sheds further light on chondrocyte-specific SOX9 expression and highlights a novel method of chondrocyte gene regulation involving a lncRNA. Summary: This study identified a chondrocyte repertoire of lncRNAs and discovered that ROCR (regulator of chondrogenesis RNA) is important for MSC chondrogenesis and cartilage gene expression by promoting the expression of SOX9.
The Journal of Allergy and Clinical Immunology | 2018
Meghan J. Acres; Florian Gothe; Andrew J. Grainger; Andrew Skelton; David Swan; Joseph D. P. Willet; Suzy Leech; Sonya Galcheva; Violeta Iotova; Sophie Hambleton; Karin R. Engelhardt
A novel homozygous missense mutation in the coiled-coil domain of STAT5B causes an immunodeficiency syndrome characterised by short stature due to GH insensitivity, immune dysregulation and hypothyroidism but currently without pulmonary disease or T-cell lymphopenia.
Annals of the Rheumatic Diseases | 2017
Laura Ridgley; Amy E. Anderson; Andrew Skelton; David Young; John D. Isaacs; Ruaidhrí J. Carmody; Arthur G. Pratt
Background Rheumatoid arthritis (RA) is a heterogeneous disease of immune dysregulation. Transcriptional profiling of circulating CD4+ T-cells identified a 12 gene signature which could distinguish untreated early arthritis patients from disease controls. This signature was enriched for STAT-3 target genes whose expression correlated with circulating IL-6 levels. We hypothesise that pre-exposure of naïve CD4+ T-cells to circulating IL-6 mediates STAT-3 activation and subsequent aberrant effector function following T-cell receptor (TCR) stimulation, providing a mechanism of antigen non-specific immune dysfunction in early RA. Materials and methods Naïve (CD45RA+) and antigen experienced (CD45RA-) CD4+ T-cells from healthy donors were cultured with IL-6 and equimolar concentrations of soluble IL-6R for 3 days to mimic chronic exposure, before being washed thoroughly and stimulated with anti-CD3/anti-CD28 for 6 days. Activation and proliferation were assessed by flow cytometry measuring cell surface markers and CFSE. RNA was extracted from cells at multiple experimental time points and global gene expression profiling undertaken using an Illumina microarray. Results Aside from downregulation of IL-6β receptor, no change in the activation of naïve CD4+ T-cells following culture with IL-6 was observed. Pre-exposure of naïve CD4+ T-cells to IL-6 and subsequent TCR stimulation enhanced proliferative capacity and cell activation, in a dose-dependent manner. Interestingly, physiological levels of IL-6, mirroring those found in the serum of RA patient’s strongly enhanced naïve CD4+ T-cell proliferation. This effect was less pronounced in memory CD4+ T-cells. In addition pre-exposure of CD4+ T-cells to IL-6 resulted in altered cytokine profiles and T-helper cell differentiation. Pre-exposure of healthy control naïve CD4+ T-cells to physiological levels of IL-6 caused significant STAT-3 activated gene induction which mirror genes previously found to distinguish RA patients from disease controls. These genes were less pronounced and unsustained in memory CD4+ T-cells. Conclusions In the proinflammatory early RA disease state our data imply that pre-exposure of cells to IL-6 causes increased proliferative capacity and activation status as well as differential gene expression, most prominently in naïve CD4+ T-cells. This highlights that cytokine ‘pre-priming’ during this critical phase may have consequences for naïve CD4+ T-cell effector function impacting the transition to disease chronicity.