Network


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

Hotspot


Dive into the research topics where Bernard J Morley is active.

Publication


Featured researches published by Bernard J Morley.


Current Biology | 2000

Autoimmune-prone mice share a promoter haplotype associated with reduced expression and function of the Fc receptor FcγRII

Nicholas R. Pritchard; Antony J. Cutler; Santiago Uribe; Steven J. Chadban; Bernard J Morley; Kenneth G. C. Smith

Human autoimmune diseases thought to arise from the combined effects of multiple susceptibility genes include systemic lupus erythematosus (SLE) and autoimmune diabetes. Well-characterised polygenic mouse models closely resembling each of these diseases exist, and genetic evidence links receptors for the Fc portion of immunoglobulin G (FcR) with their pathogenesis in mice and humans [1] [2] [3]. FcRs may be activatory or inhibitory and regulate a variety of immune and inflammatory processes [4] [5]. FcgammaRII (CD32) negatively regulates activation of cells including B cells and macrophages [6]. FcgammaRII-deficient mice are prone to immune-mediated disease [7] [8] [9]. The gene encoding FcgammaRII, Fcgr2, is contained in genetic susceptibility intervals in mouse models of SLE such as the New Zealand Black (NZB) contribution to the (NZB x New Zealand White (NZW)) F1 strain [1] [10] [11] and the BXSB strain [12], and in human SLE [1] [2] [3]. We therefore sequenced Fcgr2 and identified a haplotype defined by deletions in the Fcgr2 promoter region that is present in major SLE-prone mouse strains (NZB, BXSB, SB/Le, MRL, 129 [13]) and non-obese diabetic (NOD) mice but absent in control strains (BALB/c, C57BL/6, DBA/2, C57BL/10) and NZW mice. The autoimmune haplotype was associated with reduced cell-surface expression of FcgammaRII on macrophages and activated B cells and with hyperactive macrophages resembling those of FcgammaRII-deficient mice, and is therefore likely to play an important role in the pathogenesis of SLE and possibly diabetes.


Annals of the New York Academy of Sciences | 1997

Complement deficiency and autoimmunity.

Mark Walport; K. A. Davies; Bernard J Morley; M. Botto

There are three unexplained paradoxes that characterize the association between the complement system and systemic lupus erythematosus (SLE). The first is that complement participates in the inflammatory lesions of SLE and yet inherited homozygous deficiency of certain complement proteins is powerfully associated with the development of SLE. The second is that C lq deficiency shows the strongest association with the development of SLE and yet, in patients with SLE who do not have Clq deficiency, autoantibodies to Clq commonly develop. The third paradox is that complement deficiency is associated with an impaired primary and secondary antibody response to T lymphocyte4ependent antigens, yet SLE is characterized by high levels of autoantibodies to many intracellular and cell surface antigens. In this report, we shall review the data related to these three paradoxes and some hypotheses to explain them.


Arthritis & Rheumatism | 2000

Identification of intervals on chromosomes 1, 3, and 13 linked to the development of lupus in BXSB mice

Michelle E. K. Haywood; Mb Hogarth; Jason H. Slingsby; S. Jane Rose; Penny J. Allen; E. Mary Thompson; Michael A. Maibaum; Phillip Chandler; Kevin A. Davies; Elizabeth Simpson; Mark Walport; Bernard J Morley

OBJECTIVE To identify intervals containing systemic lupus erythematosus (SLE) susceptibility alleles in the BXSB strain of mice. METHODS We analyzed 286 (B10 x [B10 x BXSB]F1) backcross mice for a range of phenotypic traits associated with the development of SLE in BXSB mice. The mice were genotyped using 93 microsatellite markers, and the linkage of these markers to disease was studied by extreme-phenotype and quantitative trait locus analysis. RESULTS The disease phenotype in these backcross mice was less severe than that in BXSB mice. However, antinuclear antibody production was increased compared with the parental strain. We identified 4 areas of genetic linkage to disease on chromosome 1 (Bxs1-4), 1 on chromosome 3 (Bxs5), and another interval on chromosome 13 which were associated with various aspects of the phenotype. Bxs4 and Bxs5 are located in regions not previously linked to disease in other models of SLE. CONCLUSION SLE in the BXSB mouse model has a complex genetic basis and involves at least 5 distinct intervals located on chromosomes 1 and 3. There is evidence that different intervals affect particular aspects of the SLE phenotype.


Journal of Clinical Investigation | 1996

The molecular basis of hereditary complement factor I deficiency

Timothy J. Vyse; Bernard J Morley; I Bartók; El Theodoridis; Kevin A. Davies; Ad Webster; Mark Walport

The molecular basis of hereditary complement factor I deficiency is described in two pedigrees. In one pedigree, there were two factor I-deficient siblings, one of whom was asymptomatic and the other suffered from recurrent pyogenic infections. Their factor I mRNA was analyzed by reverse transcription of fibroblast RNA followed by amplification using the polymerase chain reaction. Both siblings were homozygous for the same transversion (adenine to thymine) at nucleotide 1282 in the cDNA. This mutation causes histidine-400 to be replaced by leucine. The altered histidine is a semi-conserved residue within the serine proteinase family, although no function has been ascribed to it. The proband of the second pedigree studied was found to be a compound heterozygote. One allele had the same mutation as the first family, the second allele had a donor splice site mutation that resulted in the deletion of the mRNA encoded in the fifth exon (a low-density lipoprotein receptor domain) from its transcript.


Genes and Immunity | 2006

Overlapping BXSB congenic intervals, in combination with microarray gene expression, reveal novel lupus candidate genes.

Michelle E. K. Haywood; Sj Rose; S Horswell; Mj Lees; G Fu; Mark Walport; Bernard J Morley

The BXSB mouse strain is an important model of glomerulonephritis observed in systemic lupus erythematosus (SLE). Linkage studies have successfully identified disease-susceptibility intervals; however, extracting the identity of the susceptibility gene(s) in such regions is the crucial next step. Congenic mouse strains present a defined genetic resource that is highly amenable to microarray analysis. We have performed microarray analysis using a series of chromosome 1 BXSB congenic mice with partially overlapping disease-susceptibility intervals. Simultaneous comparison of the four congenic lines allowed the identification of expression differences associated with both the initiation and progression of disease. Thus, we have identified a number of novel SLE disease gene candidates and have confirmed the identity of Ifi202 as a disease candidate in the BXSB strain. Sequencing of the promoter regions of Gas5 has revealed polymorphisms in the BXSB strain, which may account for the differential expression profile. Furthermore, the combination of the microarray results with the different phenotypes of these mice has allowed the identification of a number of expression differences that do not necessarily map to the congenic interval, but may be implicated in disease pathways.


Journal of Immunology | 2000

TNF-α, IL-4, and IFN-γ Regulate Differential Expression of P- and E-Selectin Expression by Porcine Aortic Endothelial Cells

Cj Stocker; Kl Sugars; Oa Harari; Rc Landis; Bernard J Morley; Do Haskard

P- and E-selectin are surface glycoproteins that mediate leukocyte rolling on the surface of endothelium in inflammation. We have cloned porcine P-selectin cDNA and generated a mAb, 12C5, with which to examine P-selectin expression by porcine aortic endothelial cells (PAEC) in comparison with that of E-selectin. Basal expression by PAEC of P-selectin was greater than that of E-selectin, whereas E-selectin expression was more prominently enhanced than that of P-selectin by stimulation with TNF-α or IL-1α. Both human or porcine IL-4 led to an increase in P-selectin expression, with kinetics that were delayed compared with those seen following stimulation with TNF-α or IL-1α, but IL-4 did not stimulate expression of E-selectin. When cells were stimulated with TNF-α in the presence of IL-4, we observed enhanced P-selectin expression with a parallel reduction in E-selectin expression. Finally, the increase in P-selectin expression due to human IL-4 was reduced in the presence of porcine but not human IFN-γ. These observations show that E-selectin and P-selectin expression are differentially regulated in PAEC, and that IL-4 leads to a shift in the relative surface density of the two molecules toward P-selectin. The ability of porcine IFN-γ to inhibit IL-4-induced P-selectin expression suggests that the balance between Th1 and Th2 cytokine production may determine the relative densities of the two selectins in chronic immune-mediated inflammation. Because the increased expression of P-selectin induced by human IL-4 was not inhibited by human IFN-γ, this balance may be shifted toward P-selectin expression in porcine xenografts infiltrated by human lymphocytes.


Journal of Immunology | 2004

Dissection of BXSB Lupus Phenotype Using Mice Congenic for Chromosome 1 Demonstrates That Separate Intervals Direct Different Aspects of Disease

Michelle E. K. Haywood; Nicola J. Rogers; S. Jane Rose; Joseph J. Boyle; Aileen McDermott; Joanna Rankin; Vasuky Thiruudaian; Margarita Lewis; Liliane Fossati-Jimack; Shozo Izui; Mark Walport; Bernard J Morley

To dissect the individual effects of the four non-MHC, autosomal loci (Bxs1 to Bxs4) that contribute to SLE susceptibility in BXSB mice, we generated congenic lines from chromosome 1 on a C57BL/10.YBXSB (B10.Yaa) background for the intervals (values in megabases (Mb)) Bxs1 (46.3-89.2 Mb), Bxs1/4 (20.0-65.9 Mb), Bxs1/2 (64.4-159.0 Mb), and Bxs2/3 (105.4-189.0 Mb). Glomerulonephritis, qualitatively similar to that seen in the parental BXSB strain, developed in three of these congenic strains. Early onset, severe disease was observed in B10.Yaa.BXSB-Bxs2/3 congenic mice and caused 50% mortality by 12 mo. In B10.Yaa.BXSB-Bxs1/4 mice disease progressed more slowly, resulting in 13% mortality at 12 mo. The progression of renal disease in both of these strains was correlated with the level of anti-dsDNA Abs. B10.Yaa.BXSB-Bxs1 mice, despite their genetic similarity to B10.Yaa.BXSB-Bxs1/4 mice, developed a low-grade glomerulonephritis in the absence of anti-dsDNA Abs. Thus, Bxs4 directed an increase in titer and spectrum of autoantibodies, whereas Bxs1 promoted the development of nephritis. The Bxs2 interval was linked to the production of anti-dsDNA Abs without concomitant glomerulonephritis. In contrast, the Bxs3 interval was sufficient to generate classic lupus nephritis in a nonautoimmune–prone strain. Immune phenotype differed between controls and congenics with a significant increase in B220+ cells in BXSB and B10.Yaa.BXSB-Bxs2/3, and an increase in CD4 to CD8 ratio in both BXSB and B10.Yaa.BXSB-Bxs1/4. Disease in the Bxs3 mice was delayed in comparison to the BXSB parental strain, emphasizing the necessity for multiple interactions in the production of the full BXSB phenotype.


Journal of Immunology | 2001

Autoantigen glycoprotein 70 expression is regulated by a single locus, which acts as a checkpoint for pathogenic anti-glycoprotein 70 autoantibody production and hence for the corresponding development of severe nephritis, in lupus-prone PXSB mice.

Michelle E. K. Haywood; Timothy J. Vyse; Aileen McDermott; Em Thompson; Akinori Ida; Mark Walport; Shozo Izui; Bernard J Morley

Retroviral envelope glycoprotein gp70 is present in the sera of immunologically normal and autoimmune-prone strains of mice. However, only lupus-prone mice spontaneously develop gp70-anti-gp70 immune complexes (gp70IC), and these have been implicated in the development of nephritis. We investigated the genetic factors that affect the production of both free serum gp70 and gp70IC in the lupus-prone BXSB mouse strain by analyzing (BXSB × (C57BL/10 × BXSB)F1)- and (C57BL/10 × (C57BL/10 × BXSB)F1)-backcrossed male mice. Production of gp70 mapped to a single major locus located on chromosome 13 (Bxs6) with a maximum log likelihood of the odds of 36.7 (p = 1.6 × 10−38). The level of gp70IC was highly dependent on Bxs6-related gp70 production, and high titer autoantibody production only occurred when serum gp70 levels were greater than a threshold value of ∼4.0 μg/ml. The subdivision of the (BXSB × (C57BL/10 × BXSB)F1)-backcrossed mice into those homozygous or heterozygous for Bxs6 enabled a remarkable association to be observed between high levels of gp70IC and severe nephritis in the Bxs6 homozygote population. A further mapping study in these two subgroups identified a previously unrecognized interval associated with the production of autoantibodies.


The Journal of Pathology | 2008

Control of apoptosis in autoimmunity.

Eleni Maniati; P Potter; Nicola J. Rogers; Bernard J Morley

Apoptosis and the subsequent removal of apoptotic cells underpin a healthy immune system. They are crucial for both the maintenance of self‐tolerance and the contraction of clonally expanded lymphocytes at the conclusion of immune responses. Aberrant apoptosis and the disposal of apoptotic cells is implicated in the development of both systemic and organ‐specific autoimmune disease and is a major contributing factor in disease susceptibility. Dissection of the molecular mechanisms involved in dysregulated apoptosis may reveal pathways which can be targeted for more effective therapeutic intervention. This review highlights the molecular events underlying programmed cell death and apoptotic cell uptake, and summarizes recent studies that link impaired apoptotic death to autoimmunity. Copyright


Journal of Immunology | 2009

A Defect in Marco Expression Contributes to Systemic Lupus Erythematosus Development via Failure to Clear Apoptotic Cells

Nicola J. Rogers; Mark Jeffrey Lees; L Gabriel; Eleni Maniati; Sarah Jane Rose; Paul K. Potter; Bernard J Morley

Systemic lupus erythematosus is a multisystem autoimmune disease characterized by the production of numerous antinuclear autoantibodies and inflammatory mediators. The BXSB mouse strain is an excellent model of the disease. Previous work has determined a number of important disease susceptibility intervals that have been isolated in separate congenic strains. Here, we have combined expression data from those strains with functional analyses to demonstrate that reduced expression of the innate scavenger receptor Marco (macrophage receptor with collagenous structure) is a primary event in BXSB mice, that reduced mRNA expression is mirrored at the protein level, and that this results in a significant alteration in function. We have confirmed a role for Marco in the clearance of apoptotic cells and a generalized defect in both endocytosis and phagocytosis. The failure to clear apoptotic cells has previously been linked to the development of systemic lupus erythematosus. However, the use of congenic mice with limited phenotypes in this study has enabled us to propose that in the case of Marco at least, disease results from the production of anti-dsDNA Abs.

Collaboration


Dive into the Bernard J Morley's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

L Gabriel

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eleni Maniati

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Kevin A. Davies

Brighton and Sussex Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge