Network


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

Hotspot


Dive into the research topics where Simon J. Davis is active.

Publication


Featured researches published by Simon J. Davis.


Nature | 2001

Crystal structure of the B7-1/CTLA-4 complex that inhibits human immune responses.

Carin C. Stamper; Yan Zhang; James Tobin; David V. Erbe; Shinji Ikemizu; Simon J. Davis; Mark L. Stahl; Jasbir Seehra; William S. Somers; Lidia Mosyak

Optimal immune responses require both an antigen-specific and a co-stimulatory signal. The shared ligands B7-1 and B7-2 on antigen-presenting cells deliver the co-stimulatory signal through CD28 and CTLA-4 on T cells. Signalling through CD28 augments the T-cell response, whereas CTLA-4 signalling attenuates it. Numerous animal studies and recent clinical trials indicate that manipulating these interactions holds considerable promise for immunotherapy. With the consequences of these signals well established, and details of the downstream signalling events emerging, understanding the molecular nature of these extracellular interactions becomes crucial. Here we report the crystal structure of the human CTLA-4/B7-1 co-stimulatory complex at 3.0u2009Å resolution. In contrast to other interacting cell-surface molecules, the relatively small CTLA-4/B7-1 binding interface exhibits an unusually high degree of shape complementarity. CTLA-4 forms homodimers through a newly defined interface of highly conserved residues. In the crystal lattice, CTLA-4 and B7-1 pack in a strikingly periodic arrangement in which bivalent CTLA-4 homodimers bridge bivalent B7-1 homodimers. This zipper-like oligomerization provides the structural basis for forming unusually stable signalling complexes at the T-cell surface, underscoring the importance of potent inhibitory signalling in human immune responses.


Immunology Today | 1996

The structure and ligand interactions of CD2: implications for T-cell function

Simon J. Davis; P. Anton van der Merwe

Abstract Considerable progress has recently been made in understanding the structure and ligand interactions of the T-cell antigen CD2, to the extent that CD2 is now a useful paradigm for considering the structural basis of cell-cell recognition. Here, Simon Davis and Anton van der Merwe review the new data and consider their implications for T-cell function in the context of CD2-knockout experiments.


Nature Immunology | 2006

The kinetic-segregation model: TCR triggering and beyond

Simon J. Davis; van der Merwe Pa

How the T cell receptor engages antigen is known, but not how that triggers intracellular signaling. The first direct support for a mechanism based on the spatial reorganization of signaling proteins, proposed 10 years ago and referred to as the kinetic-segregation model, is now beginning to emerge, along with indications that it may also apply to the triggering of nonclonotypic receptors. We describe here the development of the model, review new data and suggest how the model fits a broader conceptual framework for receptor triggering. We also consider the capacity of the model, versus that of other proposals, to account for the established features of TCR triggering.


Nature Methods | 2006

A rigorous experimental framework for detecting protein oligomerization using bioluminescence resonance energy transfer

John R. James; Marta I. Oliveira; Alexandre M. Carmo; Andrea Iaboni; Simon J. Davis

Bioluminescence resonance energy transfer (BRET), which relies on nonradiative energy transfer between luciferase-coupled donors and GFP-coupled acceptors, is emerging as a useful tool for analyzing the quaternary structures of cell-surface molecules. Conventional BRET analyses are generally done at maximal expression levels and single acceptor/donor ratios. We show that under these conditions substantial energy transfer arises from random interactions within the membrane. The dependence of BRET efficiency on acceptor/donor ratio at fixed surface density, or expression level at a defined acceptor/donor ratio, can nevertheless be used to correctly distinguish between well-characterized monomeric and oligomeric proteins, including a very weak dimer. The pitfalls associated with the nonrigorous treatment of BRET data are illustrated for the case of G protein–coupled receptors (GPCRs) proposed to form homophilic and/or mixed oligomers on the basis of previous, conventional BRET experiments.Please visit methagora to view and post comments on this article


Nature | 2012

TNF receptor 1 genetic risk mirrors outcome of anti-TNF therapy in multiple sclerosis.

Adam Patrick Gregory; Calliope A. Dendrou; Kathrine E. Attfield; Aiden Haghikia; Dionysia K. Xifara; Falk Butter; Gereon Poschmann; Gurman Kaur; Lydia Lambert; Oliver A. Leach; Simone Prömel; Divya Punwani; James H. Felce; Simon J. Davis; Ralf Gold; Finn C. Nielsen; Richard M. Siegel; Matthias Mann; John I. Bell; Gil McVean; Lars Fugger

Although there has been much success in identifying genetic variants associated with common diseases using genome-wide association studies (GWAS), it has been difficult to demonstrate which variants are causal and what role they have in disease. Moreover, the modest contribution that these variants make to disease risk has raised questions regarding their medical relevance. Here we have investigated a single nucleotide polymorphism (SNP) in the TNFRSF1A gene, that encodes tumour necrosis factor receptor 1 (TNFR1), which was discovered through GWAS to be associated with multiple sclerosis (MS), but not with other autoimmune conditions such as rheumatoid arthritis, psoriasis and Crohn’s disease. By analysing MS GWAS data in conjunction with the 1000 Genomes Project data we provide genetic evidence that strongly implicates this SNP, rs1800693, as the causal variant in the TNFRSF1A region. We further substantiate this through functional studies showing that the MS risk allele directs expression of a novel, soluble form of TNFR1 that can block TNF. Importantly, TNF-blocking drugs can promote onset or exacerbation of MS, but they have proven highly efficacious in the treatment of autoimmune diseases for which there is no association with rs1800693. This indicates that the clinical experience with these drugs parallels the disease association of rs1800693, and that the MS-associated TNFR1 variant mimics the effect of TNF-blocking drugs. Hence, our study demonstrates that clinical practice can be informed by comparing GWAS across common autoimmune diseases and by investigating the functional consequences of the disease-associated genetic variation.


Journal of Immunology | 2000

Distinct Pathways of Mannan-Binding Lectin (MBL)- and C1-Complex Autoactivation Revealed by Reconstitution of MBL with Recombinant MBL-Associated Serine Protease-2

Thomas Vorup-Jensen; Steen V. Petersen; Annette G. Hansen; Knud Poulsen; Wilhelm J. Schwaeble; Robert B. Sim; Kenneth B. M. Reid; Simon J. Davis; Steffen Thiel; Jens C. Jensenius

Mannan-binding lectin (MBL) plays a pivotal role in innate immunity by activating complement after binding carbohydrate moieties on pathogenic bacteria and viruses. Structural similarities shared by MBL and C1 complexes and by the MBL- and C1q-associated serine proteases, MBL-associated serine protease (MASP)-1 and MASP-2, and C1r and C1s, respectively, have led to the expectation that the pathways of complement activation by MBL and C1 complexes are likely to be very similar. We have expressed rMASP-2 and show that, whereas C1 complex autoactivation proceeds via a two-step mechanism requiring proteolytic activation of both C1r and C1s, reconstitution with MASP-2 alone is sufficient for complement activation by MBL. The results suggest that the catalytic activities of MASP-2 split between the two proteases of the C1 complex during the course of vertebrate complement evolution.


Immunity | 1999

T Cell Receptor and Coreceptor CD8αα Bind Peptide-MHC Independently and with Distinct Kinetics

Jessica R. Wyer; Benjamin E. Willcox; George F. Gao; Ulrich C. Gerth; Simon J. Davis; John I. Bell; P. Anton van der Merwe; Bent K. Jakobsen

The T cell surface glycoprotein CD8 enhances T cell antigen recognition by binding to MHC class I molecules. We show that human CD8 alphaalpha binds to the MHC class I molecule HLA-A2 with an extremely low affinity (Kd approximately 0.2 mM at 37 degrees C) and with kinetics that are between 2 and 3 orders of magnitude faster than reported for T cell receptor/peptide-MHC interactions. Furthermore, CD8 alphaalpha had no detectable effect on a T cell receptor (TCR) binding to the same peptide-MHC class I complex. These binding properties provide an explanation as to why the CD8/MHC class I interaction is unable to initiate cell-cell adhesion and how it can enhance TCR recognition without interfering with its specificity.


Nature Structural & Molecular Biology | 2003

The ligand-binding face of the semaphorins revealed by the high-resolution crystal structure of SEMA4D

Christopher Anthony Love; Karl Harlos; N Mavaddat; Simon J. Davis; David I. Stuart; E.Y. Jones; Robert M. Esnouf

Semaphorins, proteins characterized by an extracellular sema domain, regulate axon guidance, immune function and angiogenesis. The crystal structure of SEMA4D (residues 1–657) shows the sema topology to be a seven-bladed β-propeller, revealing an unexpected homology with integrins. The sema β-propeller contains a distinctive 77-residue insertion between β-strands C and D of blade 5. Blade 7 is followed by a domain common to plexins, semaphorins and integrins (PSI domain), which forms a compact cysteine knot abutting the side of the propeller, and an Ig-like domain. The top face of the β-propeller presents prominent loops characteristic of semaphorins. In addition to limited contact between the Ig-like domains, the homodimer is stabilized through extensive interactions between the top faces in a sector of the β-propeller used for heterodimerization in integrins. This face of the propeller also mediates ligand binding in integrins, and functional data for semaphorin-receptor interactions map to the equivalent surface.


Journal of Biological Chemistry | 1997

The Glycosylation of the Complement Regulatory Protein, Human Erythrocyte CD59

Pauline M. Rudd; B. Paul Morgan; Mark R. Wormald; David J. Harvey; Carmen W. van den Berg; Simon J. Davis; Michael A. J. Ferguson; Raymond A. Dwek

Human erythrocyte CD59 contains N- and O-glycans and a glycosylphosphatidylinositol (GPI) anchor, all of which have been analyzed in this study. The anchor consists principally of the minimum core glycan sequence Manα1-2Manα1-6Manα1-4GlcN-linked to a phosphatidylinositol moiety with the structure sn-1-O-alkyl(C18:0 and C18:1)-2-O-acyl(C20:4)glycerol-3-phospho-1-(2-O-palmitoyl(C16:0))myo-inositol. This structure is essentially identical to that of human erythrocyte cholinesterase (Deeg, M. A., Humphrey, D. R., Yang, S. H., Ferguson, T. R., Reinhold, V. N., and Rosenberry, T. L. (1992) J. Biol. Chem. 267, 18573-18580). This first comparison of GPI anchors from different proteins expressed in the same tissue suggests that human reticulocytes produce only one type of anchor structure. The N- and O-glycans were sequenced using a novel approach involving digestion of the total glycan pool with multiple enzyme arrays. The N-glycan pool contained families of bi-antennary complex-type structures with and without lactosamine extensions and outer arm fucose residues. The predominant O-glycans were NeuNAcα2-3Galβ1-3GalNAc and Galβ1-3[NeuNAcα2-3]GalNAc. Inspection of a molecular model of CD59, based on the NMR solution structure of the extracellular domain and the structural data from this study, suggested several roles for the glycans, including spacing and orienting CD59 on the cell surface and protecting the molecule from proteases. This work completes the initial structural analysis of CD59, providing the most complete view of any cell surface glycoprotein studied to date.


Immunological Reviews | 1998

CD2 and the nature of protein interactions mediating cell-cell recognition

Simon J. Davis; Shinji Ikemizu; Martin K. Wild; P. Anton van der Merwe

Summary: Rapid progress has recently been made in characterising the structures of leukocyte cell‐surface molecules. Detailed analyses of the structure and interactions of CD2 were the first involving a molecule that has not been directly linked to antigen recognition in the manner of antigen receptors or co‐receptors. It seems highly likely that the properties of ligand binding by CD2 are relevant to the general mechanisms of cell‐cell recognition. As an example of biological recognition, the defining characteristic of cell‐cell contact is that it involves che simultaneous interaction of hundreds, if not thousands, of molecules. Affinity and kinetic analyses of ligand binding by CD2 indicated that the protein interactions mediating cell‐cell contact, whilst highly specific, are much weaker than initially anticipated, probably due to the requirement that such contacts be easily reversible. Simultaneously, in addressing the mechanism of this mode of recognition, structural and mutational studies focussed on the role of charged residues clustered in the ligand‐binding face of CD2, yielding the concept that electrostatic complementarity, rather than surface‐shape complementarity, is the dominant feature of specific, low‐affinity protein recognition at die cell surface by CD2. The crystallographic analysis of the CD2‐binding domain of CD58 strongly supports this concept.

Collaboration


Dive into the Simon J. Davis'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

Chao Yu

University of Oxford

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge