Network


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

Hotspot


Dive into the research topics where Christopher W. Dawson is active.

Publication


Featured researches published by Christopher W. Dawson.


Journal of General Virology | 1988

Epstein—Barr Virus Gene Expression in Nasopharyngeal Carcinoma

Lawrence S. Young; Christopher W. Dawson; D. Clark; H. Rupani; Pierre Busson; Thomas Tursz; A. Johnson; Alan B. Rickinson

Epstein-Barr virus (EBV), an agent with growth transforming potential for human B cells, is associated with certain B cell lymphomas in man and also with an epithelial tumour, undifferentiated nasopharyngeal carcinoma (NPC). Since B cell growth transformation is associated with the constitutive expression of a small number of EBV-coded latent proteins, the nuclear antigens EBNA 1, EBNA 2, EBNA 3 and EBNA-LP and the latent membrane protein (LMP), the present work sought to determine whether this same pattern of virus gene expression occurred in NPC. Tumour biopsies were taken from NPC patients from three areas of differing tumour incidence (Kenya, Algeria, Britain) and immediately snap-frozen, as were biopsies of non-EBV-related carcinomas for controls. Immunoblotting of PAGE-separated proteins with selected human sera identified 24 NPC biopsies clearly expressing EBNA 1. When the analysis was extended using selected human sera with antibodies against the other EBNAs, there was no detectable expression of EBNA 2, EBNA 3 or EBNA-LP in any of these 24 biopsies; their EBNA 2-negative status was confirmed using a monoclonal antibody (MAb) PE2 which was reactive in immunoblotting and in immunoprecipitation with EBNA 2A and EBNA 2B proteins. Similar experiments with two different LMP-specific MAbs, CS1 to 4 and S12, revealed heterogeneity between NPC biopsies; 9/24 biopsies were demonstrably LMP-positive, the degree of expression varying considerably between individual tumours in a manner which was not related to the level of EBNA 1 expression. None of the 24 NPC biopsies expressed detectable amounts of EBV lytic cycle antigens. A nude mouse-passaged NPC cell line, C15, likewise expressed EBNA 1 and LMP but none of the other EBV latent proteins nor lytic cycle antigens. This work identifies a novel type of EBV-cell interaction in NPC cells which is distinct from that seen in in vitro transformed B cell lines and from that seen to date in EBV-positive B cell lymphomas.


Journal of Biological Chemistry | 1999

Activation of the p38 mitogen-activated protein kinase pathway by Epstein-Barr virus-encoded latent membrane protein 1 coregulates interleukin-6 and interleukin-8 production.

Aristides G. Eliopoulos; Neil J. Gallagher; Sarah M. S. Blake; Christopher W. Dawson; Lawrence S. Young

The Epstein-Barr virus-encoded latent membrane protein 1 (LMP1) is a pleiotropic protein the activities of which include effects on gene expression and cell transformation, growth, and death. LMP1 has been shown to induce nuclear factor (NF)-κB and c-Jun NH2-terminal kinase/AP-1 activities in target cells, and in this study we demonstrate that LMP1 also engages the p38 mitogen-activated protein kinase cascade, leading to activation of the transcription factor ATF2. Mutational analysis of the LMP1 cytoplasmic COOH terminus revealed that p38 activation occurs from both the tumor necrosis factor receptor-associated factor (TRAF)-interacting, membrane-proximal COOH-terminal activating region (CTAR)1 domain (amino acids 186–231) and the extreme tumor necrosis factor receptor-associated death domain (TRADD) binding CTAR2 region (amino acids 351–386). Because LMP1 also engages signaling on the NF-κB axis through CTAR1 and CTAR2, we have examined whether these two pathways are overlapping or independent. We have found that inhibition of p38 by the highly specific inhibitor SB203580 did not affect NF-κB binding activity. Conversely, although the metabolic inhibitor D609 blocked NF-κB activation, it did not impair the ability of LMP1 to signal on the p38 axis, suggesting that these two LMP1-mediated pathways are primarily independent. Divergence of signals must, however, occur downstream of TRAF2 as a dominant negative TRAF2 mutant that blocks LMP1-induced NF-κB activation also inhibited p38 signaling. In addition, we have found that p38 inhibition significantly impaired LMP1-mediated interleukin-6 and -8 expression. Thus, p38 may play a significant cooperative role in regulating at least some of the pleiotropic activities of LMP1.


Journal of Biological Chemistry | 2003

Epstein-Barr Virus Latent Membrane Protein 1 (LMP1) Activates the Phosphatidylinositol 3-Kinase/Akt Pathway to Promote Cell Survival and Induce Actin Filament Remodeling

Christopher W. Dawson; Giorgos Tramountanis; Aristides G. Eliopoulos; Lawrence S. Young

The Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) is an integral membrane protein that functions as a constitutively activated member of the tumor necrosis factor receptor family. Whereas LMP1 has been shown to activate the NF-κB and mitogen-activated protein kinase pathways, these effects alone are unable to account for the profound oncogenic properties of LMP1. Here we show that LMP1 can activate phosphatidylinositol 3-kinase (PI3K), a lipid kinase responsible for activating a diverse range of cellular processes in response to extracellular stimuli. LMP1 was found to stimulate PI3K activity inducing phosphorylation and subsequent activation of Akt, a downstream target of PI3K responsible for promoting cell survival. Treatment of LMP1-expressing cells with the PI3K inhibitor LY294002 resulted in decreased cell survival. The tumor necrosis factor receptor-associated factor-binding domain of LMP1 was found to be responsible for PI3K activation. The ability of LMP1 to induce actin stress-fiber formation, a Rho GTPase-mediated phenomenon, was also dependent on PI3K activation. These data implicate PI3K activation in many of the LMP1-induced phenotypic effects associated with transformation and suggests that this pathway contributes both to the oncogenicity of this molecule and its role in the establishment of persistent EBV infection.


Oncogene | 1997

Epstein – Barr virus-encoded LMP1 and CD40 mediate IL-6 production in epithelial cells via an NF-κB pathway involving TNF receptor-associated factors

Aristides G. Eliopoulos; Maria Stack; Christopher W. Dawson; Kenneth M Kaye; Liz Hodgkin; Sim Sihota; Martin Rowe; Lawrence S. Young

Expression of the Epstein – Barr virus (EBV) transforming LMP1 in B cells activates the transcription factor NF-κB and induces phenotypic changes through two distinct domains in the cytoplasmic C-terminus of the protein. The aa 187 – 231 domain of LMP1, which is important for growth transformation, binds tumour necrosis factor (TNF) receptor associated factor (TRAF) 1 and TRAF3 and this interaction mediates subsequent signalling events. The TRAFs also associate with CD40, a member of the TNFR family, which upon ligation activates NF-κB and induces phenotypic changes similar to those mediated by LMP1. This study demonstrates that LMP1 expression in carcinoma cell lines and SV40-transformed keratinocytes results in induction of the pleiotropic cytokine interleukin 6 (IL6), an effect which is also observed upon CD40 ligation. The mechanism by which either LMP1 expression or CD40 ligation induces IL6 production was found to be NF-κB-dependent. Mutational analysis identified domains in the C-terminus of LMP1 which are important for NF-κB activation and IL6 secretion. LMP1 and CD40 share a common PxQxT core TRAF binding motif and mutations in or adjacent to this sequence impaired the ability of LMP1 or CD40 to induce NF-κB activation and IL6 secretion. The importance of TRAF interactions in mediating these effects was confirmed using dominant negative TRAF2 and TRAF3 mutants which also identified differences in the signalling events mediated by the two NF-κB activating domains of LMP1. A20, an anti-apoptotic protein which interacts with TRAF2 and blocks CD40-mediated NF-κB activity, also blocked NF-κB and IL6 secretion in LMP1-transfected epithelial cells. These results suggest that LMP1 regulates IL6 production in epithelial cells in a manner similar to CD40 ligation and implicate TRAFs as common mediators in the transduction of signals generated via the CD40 and LMP1 pathways. As a role for IL6 in regulating epithelial cell growth has previously been suggested, the control of IL6 secretion via the CD40 and LMP1 pathways may have implications for the growth of both normal and transformed epithelial cells.


Molecular Cancer | 2010

Epstein-Barr virus-encoded EBNA1 inhibits the canonical NF-κB pathway in carcinoma cells by inhibiting IKK phosphorylation

Robert Valentine; Christopher W. Dawson; Chunfang Hu; Khilan M Shah; Thomas J Owen; Kathryn L. Date; Sonia P Maia; Jianyong Shao; John R. Arrand; Lawrence S. Young; John D. O'Neil

BackgroundThe Epstein-Barr virus (EBV)-encoded EBNA1 protein is expressed in all EBV-associated tumours, including undifferentiated nasopharyngeal carcinoma (NPC), where it is indispensable for viral replication, genome maintenance and viral gene expression. EBNA1s transcription factor-like functions also extend to influencing the expression of cellular genes involved in pathways commonly dysregulated during oncogenesis, including elevation of AP-1 activity in NPC cell lines resulting in enhancement of angiogenesis in vitro. In this study we sought to extend these observations by examining the role of EBNA1 upon another pathway commonly deregulated during carcinogenesis; namely NF-κB.ResultsIn this report we demonstrate that EBNA1 inhibits the canonical NF-κB pathway in carcinoma lines by inhibiting the phosphorylation of IKKα/β. In agreement with this observation we find a reduction in the phosphorylation of IκBα and reduced phosphorylation and nuclear translocation of p65, resulting in a reduction in the amount of p65 in nuclear NF-κB complexes. Similar effects were also found in carcinoma lines infected with recombinant EBV and in the EBV-positive NPC-derived cell line C666-1. Inhibition of NF-κB was dependent upon regions of EBNA1 essential for gene transactivation whilst the interaction with the deubiquitinating enzyme, USP7, was entirely dispensable. Furthermore, in agreement with EBNA1 inhibiting p65 NF-κB we demonstrate that p65 was exclusively cytoplasmic in 11 out of 11 NPC tumours studied.ConclusionsInhibition of p65 NF-κB in murine and human epidermis results in tissue hyperplasia and the development of squamous cell carcinoma. In line with this, p65 knockout fibroblasts have a transformed phenotype. Inhibition of p65 NF-κB by EBNA1 may therefore contribute to the development of NPC by inducing tissue hyperplasia. Furthermore, inhibition of NF-κB is employed by viruses as an immune evasion strategy which is also closely linked to oncogenesis during persistent viral infection. Our findings therefore further implicate EBNA1 in playing an important role in the pathogenesis of NPC.


Oncogene | 2002

Tumour necrosis factor-α in Barrett's oesophagus: a potential novel mechanism of action

Chris Tselepis; Ian Perry; Christopher W. Dawson; Rob Hardy; S. Jane Darnton; Chris McConkey; Rob C Stuart; Nicholas A. Wright; Rebecca Harrison; Janusz Antoni Z Jankowski

Barretts metaplasia (BM) is an early lesion in the progression from oesophageal inflammation through dysplasia to the development of Barretts adenocarcinoma (BA). Previous work indicates that BM and BA are associated with reduced E-cadherin expression and increased cytoplasmic/nuclear pools of its associated protein β-catenin. β-catenin participates in Wnt signalling and activates oncogene transcription by complexing with T-cells factors (TCF). One such oncogene is c-myc. We have previously shown that TNF-α can down-regulate E-cadherin expression. Here, we assess TNF-α expression in Barretts metaplasia and examine if TNF-α can promote β-catenin mediated transcription of oncogenes in a gastrointestinal model system. Employing immunohistochemistry and Western blot analysis of oesophageal tissue, epithelial expression of TNF-α increases with progression along the metaplasia–dysplasia–carcinoma sequence (P<0.001). β-catenin mediated transcription was then assessed in TNF-α stimulated cell lines using the TOPFLASH reporter system whilst c-myc expression was assessed by real time PCR. In a columnar intestinal cell model, TNF-α induces c-myc expression which is induced via β-catenin mediated transcription (P<0.05). This β-catenin mediated transcription is independent of NF-κB activation. Thus, TNF-α is up-regulated in the progression of Barretts oesophagus and β-catenin mediated transcription of c-myc is a novel pathway whereby elevated levels of TNF-α may lead to oncogene transcription and altered biology in gastrointestinal epithelia and metaplasia.


Seminars in Cancer Biology | 2002

The significance of LMP1 expression in nasopharyngeal carcinoma

Sai Wah Tsao; Giorgos Tramoutanis; Christopher W. Dawson; Angela K.F Lo; Dolly P. Huang

The Epstein-Barr virus (EBV)-encoded latent membrane protein 1 (LMP1) is a key effector of EBV-mediated B cell transformation. LMP1 displays potent oncogenic properties in rodent fibroblasts, and induces a wide range of effects in B cells and epithelial cells. LMP1 functions as a constitutively active tumor necrosis factor receptor (TNFR) engaging a multitude of signaling pathways that include NF-kappaB, the mitogen-activated protein kinases (MAPKs), JNK, p38, the JAK/STAT pathway and, more recently, the small Rho GTPases. The constitutive activation of these signaling cascades explains LMP1s ability to induce such a diverse array of morphological and phenotypic effects in cells and provides an insight into how LMP1 may induce cell transformation. The frequent expression of LMP1 in undifferentiated nasopharyngeal carcinoma (NPC) points to a role for this viral oncoprotein as a key effector molecule in NPC pathogenesis.


Immunology Today | 1998

CD40 and epithelial cells: across the great divide

Lawrence S. Young; Aristides G. Eliopoulos; Neil J. Gallagher; Christopher W. Dawson

The widespread expression of CD40 in normal epithelial cells and carcinoma cells suggests that this receptor has important, additional influences beyond that of regulating immune responses. Here, Lawrence Young and colleagues discuss the effect of CD40 ligation on epithelial cells and consider the role of this pathway in the pathogenesis and treatment of carcinomas.


The Journal of Pathology | 2005

Constitutive activation of phosphatidyl-inositide 3 kinase contributes to the survival of Hodgkin's lymphoma cells through a mechanism involving Akt kinase and mTOR

Amanda Dutton; Gary M. Reynolds; Christopher W. Dawson; Lawrence S. Young; Paul G. Murray

The molecular mechanisms underlying the pathogenesis of the malignant Hodgkins/Reed–Sternberg (HRS) cells of Hodgkins lymphoma (HL) are largely unknown. This study investigates the contribution of phosphatidyl‐inositide 3 kinase (PI3‐kinase) and demonstrates that Akt, a substrate of PI3‐kinase, is constitutively activated in HL‐derived cell lines. Several downstream effectors of Akt signalling, including glycogen synthase kinase 3 (GSK‐3) α and β and mTOR substrates 4E‐BP1 and p70 S6 kinase, were also phosphorylated in HL cells. The mTOR inhibitor, rapamycin, inhibited phosphorylation of these proteins. Furthermore, LY294002 inhibited phosphorylation of p70 S6 kinase and 4E‐BP1, suggesting that the phosphorylation of p70 S6 kinase and 4E‐BP1 in HL cells is PI3‐kinase dependent. Importantly, HRS cells of primary tumour samples not only expressed high levels of activated Akt but also displayed phosphorylation of downstream targets of Akt activation including GSK‐3, 4E‐BP1, and p70 S6 Kinase. Inhibition of PI3‐kinase and mTOR showed only modest effects on cell survival at the lower serum concentrations. However, rapamycin and doxorubicin acted synergistically to reduce HL cell survival. A combination of rapamycin and chemotherapy should be investigated in the treatment of HL. Copyright


Journal of Immunology | 2000

CTL Control of EBV in Nasopharyngeal Carcinoma (NPC): EBV-Specific CTL Responses in the Blood and Tumors of NPC Patients and the Antigen-Processing Function of the Tumor Cells

Steven P. Lee; Anthony T.C. Chan; Siu Tim Cheung; Wendy A. Thomas; Debbie Croom-Carter; Christopher W. Dawson; Ching Hwa Tsai; Sing Fai Leung; Philip J. Johnson; Dolly P. Huang

Undifferentiated nasopharyngeal carcinoma (NPC) is latently infected with EBV and expresses a restricted number of viral proteins. Studies in healthy virus carriers have demonstrated that at least some of these proteins can act as targets for HLA class I-restricted CTLs. Therefore we have explored the possibility of a CTL-based therapy for NPC by characterizing EBV-specific CTL responses in 10 newly diagnosed NPC cases and 21 healthy virus carriers from Southeast Asia. Using the autologous EBV-transformed lymphoblastoid cell line, virus-specific CTL were reactivated in vitro from PBMC, cloned, and screened for cytotoxicity against target cells expressing individual EBV proteins from recombinant vaccinia vectors. EBV-specific CTLs were identified in 6 of 10 patients and 14 of 21 controls and mainly targeted the EBV nuclear Ag 3 (EBNA3) family of viral latent proteins. However, in 3 of 10 patients and 11 of 21 controls, CTLs specific for the NPC-associated protein LMP2 were also detected, albeit at low frequency. EBV-specific CTLs were detected in tumor biopsy material obtained from 3 of 6 of the patients, indicating that functional CTL are present at the tumor site, but none was specific for tumor-associated viral proteins. To assess the Ag-presenting function in NPC we studied two NPC-derived cell lines (C15 and c666.1) and demonstrated that both were capable of processing and presenting endogenously synthesized protein to HLA class I-restricted CTL clones. Overall, our data provide a sound theoretical basis for therapeutic strategies that aim to boost or elicit LMP2-specific CTL responses in NPC patients.

Collaboration


Dive into the Christopher W. Dawson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wenbin Wei

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar

John R. Arrand

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar

Ciaran Woodman

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar

Paul G. Murray

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar

Kwok Wai Lo

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John D. O'Neil

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge