Network


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

Hotspot


Dive into the research topics where Annika Burleigh is active.

Publication


Featured researches published by Annika Burleigh.


Arthritis & Rheumatism | 2009

Fibroblast growth factor 2 is an intrinsic chondroprotective agent that suppresses ADAMTS‐5 and delays cartilage degradation in murine osteoarthritis

Shi-Lu Chia; Yasunobu Sawaji; Annika Burleigh; Celia McLean; Julia J. Inglis; Jeremy Saklatvala; Tonia L. Vincent

OBJECTIVE We have previously identified in articular cartilage an abundant pool of the heparin-binding growth factor, fibroblast growth factor 2 (FGF-2), which is bound to the pericellular matrix heparan sulfate proteoglycan, perlecan. This pool of FGF-2 activates chondrocytes upon tissue loading and is released following mechanical injury. In vitro, FGF-2 suppresses interleukin-1-driven aggrecanase activity in human cartilage explants, suggesting a chondroprotective role in vivo. We undertook this study to investigate the in vivo role of FGF-2 in murine cartilage. METHODS Basal characteristics of the articular cartilage of Fgf2(-/-) and Fgf2(+/+) mice were determined by histomorphometry, nanoindentation, and quantitative reverse transcriptase-polymerase chain reaction. The articular cartilage was graded histologically in aged mice as well as in mice in which osteoarthritis (OA) had been induced by surgical destabilization of the medial meniscus. RNA was extracted from the joints of Fgf2(-/-) and Fgf2(+/+) mice following surgery and quantitatively assessed for key regulatory molecules. The effect of subcutaneous administration of recombinant FGF-2 on OA progression was assessed in Fgf2(-/-) mice. RESULTS Fgf2(-/-) mice were morphologically indistinguishable from wild-type (WT) animals up to age 12 weeks; the cartilage thickness and proteoglycan staining were equivalent, as was the mechanical integrity of the matrix. However, Fgf2(-/-) mice exhibited accelerated spontaneous and surgically induced OA. Surgically induced OA in Fgf2(-/-) mice was suppressed to levels in WT mice by subcutaneous administration of recombinant FGF-2. Increased disease in Fgf2(-/-) mice was associated with increased expression of messenger RNA of Adamts5, the key murine aggrecanase. CONCLUSION These data identify FGF-2 as a novel endogenous chondroprotective agent in articular cartilage.


Arthritis & Rheumatism | 2012

Joint immobilization prevents murine osteoarthritis and reveals the highly mechanosensitive nature of protease expression in vivo

Annika Burleigh; A Chanalaris; Matthew D. Gardiner; C. Driscoll; Olga Boruc; Jeremy Saklatvala; Tonia L. Vincent

OBJECTIVE Mechanical joint loading is critical for the development of osteoarthritis (OA). Although once regarded as a disease of cartilage attrition, OA is now known to be controlled by the expression and activity of key proteases, such as ADAMTS-5, that drive matrix degradation. This study was undertaken to investigate the link between protease expression and mechanical joint loading in vivo. METHODS We performed a microarray analysis of genes expressed in the whole joint following surgical induction of murine OA (by cutting the medial meniscotibial ligament). Gene expression changes were validated by reverse transcriptase-polymerase chain reaction in whole joints and microdissected tissues of the joint, including the articular cartilage, meniscus, and epiphysis. Following surgery, mouse joints were immobilized, either by prolonged anesthesia or by sciatic neurectomy. RESULTS Many genes were regulated in the whole joint within 6 hours of surgical induction of OA in the mouse. These included Arg1, Ccl2, Il6, Tsg6, Mmp3, Il1b, Adamts5, Adamts4, and Adamts1. All of these were significantly regulated in the articular cartilage. When joints were immobilized by prolonged anesthesia, regulation of the vast majority of genes was abrogated. When joints were immobilized by sciatic neurectomy, regulation of selected genes was abrogated, and OA was prevented up to 12 weeks postsurgery. CONCLUSION These findings indicate that gene expression in the mouse joint following the induction of OA is rapid and highly mechanosensitive. Regulated genes include the known pathogenic protease ADAMTS-5. Targeting the mechanosensing mechanisms of joint tissue may offer new strategies for disease modification.


Pain | 2010

Treatment of murine osteoarthritis with TrkAd5 reveals a pivotal role for nerve growth factor in non-inflammatory joint pain.

Kay McNamee; Annika Burleigh; Luke L. Gompels; Marc Feldmann; Shelley J Allen; Richard O. Williams; David Dawbarn; Tonia L. Vincent; Julia J. Inglis

&NA; The origin of pain in osteoarthritis is poorly understood, but it is generally thought to arise from inflammation within the innervated structures of the joint, such as the synovium, capsule and bone. We investigated the role of nerve growth factor (NGF) in pain development in murine OA, and the analgesic efficacy of the soluble NGF receptor, TrkAD5. OA was induced in mice by destabilisation of the medial meniscus and pain was assessed by measuring hind‐limb weight distribution. RNA was extracted from joints, and NGF and TNF expressions were quantified. The effect of tumour necrosis factor (TNF) and neutrophil blockade on NGF expression and pain were also assessed. NGF was induced in the joints during both post‐operative (day 3) and OA (16 weeks) pain, but not in the non‐painful stage of disease (8 weeks post‐surgery). TrkAd5 was highly effective at suppressing pain in both phases. Induction of NGF in the post‐operative phase of pain was TNF‐dependent as anti‐TNF reduced NGF expression in the joint and abrogated pain. However, TNF was not regulated in the late OA joints, and pain was not affected by anti‐TNF therapy. Fucoidan, by suppressing cellular infiltration into the joint, was able to suppress post‐operative, but not late OA pain. These results indicate that NGF is an important mediator of OA pain and that TrkAd5 represents a potent novel analgesic in this condition. They also suggest that, unlike post‐operative pain, induction of pain in OA may not necessarily be driven by classical inflammatory processes.


Arthritis & Rheumatism | 2013

Fibroblast Growth Factor 2 Drives Changes in Gene Expression Following Injury to Murine Cartilage In Vitro and In Vivo

Ka-Wing Chong; A Chanalaris; Annika Burleigh; Huilin Jin; F E Watt; Jeremy Saklatvala; Tonia L. Vincent

Objective The articular cartilage is known to be highly mechanosensitive, and a number of mechanosensing mechanisms have been proposed as mediators of the cellular responses to altered mechanical load. These pathways are likely to be important in tissue homeostasis as well as in the pathogenesis of osteoarthritis. One important injury-activated pathway involves the release of pericellular fibroblast growth factor 2 (FGF-2) from the articular cartilage. Using a novel model of murine cartilage injury and surgically destabilized joints in mice, we examined the extent to which FGF-2 contributes to the cellular gene response to injury. Methods Femoral epiphyses from 5-week-old wild-type mice were avulsed and cultured in serum-free medium. Explant lysates were Western blotted for phospho-JNK, phospho-p38, and phospho-ERK or were fixed for immunohistochemical analysis of the nuclear translocation of p65 (indicative of NF-κB activation). RNA was extracted from injured explants, rested explants that had been stimulated with recombinant FGF-2 or FGF-18, or whole joints from either wild-type mice or FGF-2−/− mice. Reverse transcription–polymerase chain reaction was performed to examine a number of inflammatory response genes that had previously been identified in a microarray analysis. Results Murine cartilage avulsion injury resulted in rapid activation of the 3 MAP kinase pathways as well as NF-κB. Almost all genes identified in murine joints following surgical destabilization were also regulated in cartilage explants upon injury. Many of these genes, including those for activin A (Inhba), tumor necrosis factor–stimulated gene 6 (Tnfaip6), matrix metalloproteinase 19 (Mmp19), tissue inhibitor of metalloproteinases 1 (Timp1), and podoplanin (Pdpn), were significantly FGF-2 dependent following injury to cartilage in vitro and to joint tissues in vivo. Conclusion FGF-2–dependent gene expression occurs in vitro and in vivo in response to cartilage/joint injury in mice.


Osteoarthritis and Cartilage | 2015

Transcriptional analysis of micro-dissected articular cartilage in post-traumatic murine osteoarthritis.

Matthew D. Gardiner; Tonia L. Vincent; C. Driscoll; Annika Burleigh; George Bou-Gharios; Jeremy Saklatvala; Hiroyuki Nagase; A Chanalaris

Summary Objective Identify gene changes in articular cartilage of the medial tibial plateau (MTP) at 2, 4 and 8 weeks after destabilisation of the medial meniscus (DMM) in mice. Compare our data with previously published datasets to ascertain dysregulated pathways and genes in osteoarthritis (OA). Design RNA was extracted from the ipsilateral and contralateral MTP cartilage, amplified, labelled and hybridized on Illumina WGv2 microarrays. Results were confirmed by real-time polymerase chain reaction (PCR) for selected genes. Results Transcriptional analysis and network reconstruction revealed changes in extracellular matrix and cytoskeletal genes induced by DMM. TGFβ signalling pathway and complement and coagulation cascade genes were regulated at 2 weeks. Fibronectin (Fn1) is a hub in a reconstructed network at 2 weeks. Regulated genes decrease over time. By 8 weeks fibromodulin (Fmod) and tenascin N (Tnn) are the only dysregulated genes present in the DMM operated knees. Comparison with human and rodent published gene sets identified genes overlapping between our array and eight other studies. Conclusions Cartilage contributes a minute percentage to the RNA extracted from the whole joint (<0.2%), yet is sensitive to changes in gene expression post-DMM. The post-DMM transcriptional reprogramming wanes over time dissipating by 8 weeks. Common pathways between published gene sets include focal adhesion, regulation of actin cytoskeleton and TGFβ. Common genes include Jagged 1 (Jag1), Tetraspanin 2 (Tspan2), neuroblastoma, suppression of tumourigenicity 1 (Nbl1) and N-myc downstream regulated gene 2 (Ndrg2). The concomitant genes and pathways we identify may warrant further investigation as biomarkers or modulators of OA.


Arthritis Research & Therapy | 2012

Pathogenic protease expression in murine OA is critically dependent upon mechanical joint loading

Annika Burleigh; A Chanalaris; Jeremy Saklatvala; Tonia L. Vincent


Osteoarthritis and Cartilage | 2017

TSG-6 is in Part Responsible for FGF2-dependent Chondroprotection in Murine Osteoarthritis

S. Lympany; J. Zarebska; A Chanalaris; Annika Burleigh; Tonia L. Vincent


Osteoarthritis and Cartilage | 2014

IL18 is secreted upon joint injury and is a key pathogenic cytokine in murine osteoarthritis

Tonia L. Vincent; A Chanalaris; J. Zarebska; S Bullers; Bryony Stott; Annika Burleigh; E Paterson; F E Watt; Jeremy Saklatvala


Osteoarthritis and Cartilage | 2012

The molecular response to acute joint destabilisation is altered in female compared to male mice

C. Driscoll; Annika Burleigh; A Chanalaris; J Saklatvala; Tonia L. Vincent


Rheumatology | 2010

Concurrent Oral 11 – Osteoarthritis [OP73–OP78]OP73. Mechanical Load Drives Inflammatory Gene Expression and Disease in Murine OA

Annika Burleigh; Jeremy Saklatvala; Tonia L. Vincent; Evin Sowden; David Kynaston; Madhurima Rai; Elliot Yates; Winston Kim; A. J. Freemont; Kirsten M. White; Anushka Soni; A Kiran; Lyndsey M. Goulston; Deborah J. Hart; Tim D. Spector; Kassim Javaid; N K Arden; Rachel Duncan; George Peat; Elaine Thomas; Elaine M. Hay; Peter Croft; Ajay Abraham; Iain Goff; Mark S. Pearce; Roger M. Francis; Fraser Birrell; Shea Palmer; Melissa Domaille; Fiona Cramp

Collaboration


Dive into the Annika Burleigh's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

A Kiran

University of Oxford

View shared research outputs
Top Co-Authors

Avatar

A. J. Freemont

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge