Network


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

Hotspot


Dive into the research topics where Diane Scott is active.

Publication


Featured researches published by Diane Scott.


Immunity | 2000

Dendritic cells permit identification of genes encoding MHC class II-restricted epitopes of transplantation antigens.

Diane Scott; Caroline Addey; Pamela Ellis; Edward James; Michael J. Mitchell; Noémie Saut; Stipo Jurcevic; Elizabeth Simpson

Minor or histocompatibility (H) antigens are recognized by CD4+ and CD8+ T lymphocytes as short polymorphic peptides associated with MHC molecules. They are the targets of graft versus host and graft versus leukemia responses following bone marrow transplantation between HLA-identical siblings. Several genes encoding class I-restricted minor H epitopes have been identified, but approaches used for these have proved difficult to adapt for cloning class II-restricted minor H genes. We have combined the unique antigen-presenting properties of dendritic cells and high levels of episomal expression following transfection of COS cells to identify a Y chromosome gene encoding two HY peptide epitopes, HYAb and HYEk.


Transplant Immunology | 2002

Minor H antigens: genes and peptides.

Elizabeth Simpson; Diane Scott; E. James; Giovanna Lombardi; K. Cwynarski; Francesco Dazzi; M. Millrain; P.J. Dyson

In this review, we describe the evidence from which the existence of non-MHC histocompatibility (H) antigens was deduced, the clinical setting of bone marrow transplantation in which they are important targets for T cell responses, and the current understanding of their molecular identity. We list the peptide epitopes, their MHC restriction molecules and the genes encoding them, of the human and murine minor H antigens now identified at the molecular level. Identification of the peptide epitopes allows T cell responses to these antigens following transplantation of MHC-matched, minor H-mismatched tissues to be enumerated using tetramers and elispot assays. This will facilitate analysis of correlations with HVG, GVH and GVL reactions in vivo. The potential to use minor H peptides to modulate in vivo responses to minor H antigens is discussed. Factors controlling immunodominance of T cell responses to one or a few of many potential minor H antigens remain to be elucidated but are important for making predictions of in vivo HVG, GVH and GVL responses and tailoring therapy after HLA-matched BMT and DLI.


Journal of Immunology | 2001

Examination of HY Response: T Cell Expansion, Immunodominance, and Cross-Priming Revealed by HY Tetramer Analysis

Millrain Mm; P. Chandler; Francesco Dazzi; Diane Scott; E. Simpson; Dyson Pj

We have applied MHC class I tetramers representing the two H2b MHC class I-restricted epitopes of the mouse male-specific minor transplantation Ag, HY, to directly determine the extent of expansion and immunodominance within the CD8+ T cell compartment following exposure to male tissue. Immunization with male bone marrow (BM), spleen, dendritic cells (DCs) and by skin graft led to rapid expansion of both specificities occupying up to >20% of the CD8+ T cell pool. At a high dose, whole BM or spleen were found to be more effective at stimulating the response than BM-derived DCs. In vivo, immunodominance within the responding cell population was only observed following chronic Ag stimulation, whereas epitope immunodominance was established rapidly following in vitro restimulation. Peptide affinity for the restricting MHC molecule was greater for the immunodominant epitope, suggesting that this might be a factor in the emergence of immunodominance. Using tetramers, we were able to directly visualize the cross-primed CD8+ HY response, but we did not find it to be the principal route for MHC class I presentation. Immunization with female spleen or DCs coated with the full complement of defined HY peptides, including the Ab-restricted CD4+ Th cell determinant, failed to induce tetramer-reactive cells.


Nature Communications | 2014

Integrin CD11b positively regulates TLR4-induced signalling pathways in dendritic cells but not in macrophages

Guang Sheng Ling; Jason Bennett; Kevin J. Woollard; Marta Szajna; Liliane Fossati-Jimack; Philip R. Taylor; Diane Scott; Guido Franzoso; H. Terence Cook; Marina Botto

Tuned and distinct responses of macrophages and dendritic cells to Toll-like receptor 4 (TLR4) activation induced by lipopolysaccharide (LPS) underpin the balance between innate and adaptive immunity. However, the molecule(s) that confer these cell-type-specific LPS-induced effects remain poorly understood. Here we report that the integrin αM (CD11b) positively regulates LPS-induced signalling pathways selectively in myeloid dendritic cells but not in macrophages. In dendritic cells, which express lower levels of CD14 and TLR4 than macrophages, CD11b promotes MyD88-dependent and MyD88-independent signalling pathways. In particular, in dendritic cells CD11b facilitates LPS-induced TLR4 endocytosis and is required for the subsequent signalling in the endosomes. Consistent with this, CD11b deficiency dampens dendritic cell-mediated TLR4-triggered responses in vivo leading to impaired T-cell activation. Thus, by modulating the trafficking and signalling functions of TLR4 in a cell-type-specific manner CD11b fine tunes the balance between adaptive and innate immune responses initiated by LPS.


European Journal of Immunology | 2002

CD4+CD25+ T cells as immunoregulatory T cells in vitro.

Jian-Guo Chai; Julia Y. S. Tsang; Robert I. Lechler; Elizabeth Simpson; Julian Dyson; Diane Scott

We have further characterized the in vitro phenotype and function of anergic and suppressive CD4+25+ T cells. Following TCR ligation, DO.11.10 CD4+25+ T cells suppress the activation of OT‐1 CD8+25– T cells in an antigen nonspecific manner. Although suppression was seen when using a mixture of APC from both parental strains, it was very much more marked when using F1 APC. APC pretreated with, and then separated from CD4+25+ T cells did not have diminished T cell costimulatory function, suggesting that APC are not the direct targets of CD4+25+ T cell regulation. CTLA‐4 blockade failed to abrogate suppression by CD4+25+ T cells in mixing experiments. Although CD4+25+ T cells failed to respond following cross‐linking of TCR, they could be induced to proliferate following the addition of exogenous IL‐2, allowing the generation of a T cell line from CD4+25+ T cells. After the first in vitro restimulation, CD4+25+ T cells were still anergic and suppressive following TCR engagement. However, after three rounds of restimulation, their anergic and suppressive status was abrogated.


Journal of Immunology | 2008

In Vitro Expansion Improves In Vivo Regulation by CD4+CD25+ Regulatory T Cells

Jian-Guo Chai; David Coe; Daxin Chen; Elizabeth Simpson; Julian Dyson; Diane Scott

CD4+CD25+ T regulatory cells (Tregs) can actively suppress immune responses and thus have substantial therapeutical potential. Clinical application is, however, frustrated by their scarcity, anergic status, and lack of defined specificity. We found that a single injection of a small number of expanded but not fresh HY-specific Tregs protected syngeneic male skin grafts from rejection by immune-competent recipients. The expanded Tregs were predominantly located in the grafts and graft-draining lymph nodes. In vitro expanded Tregs displayed a phenotype of CD25highCD4lowFoxp3+CTLA4+, and also up-regulated IL10 and TGFβ while down-regulating IFN-γ, GM-CSF, IL5, and TNF-α production. Furthermore, expanded Tregs appeared to express a reduced level of Foxp3, which could be prevented by adding TGFβ to the culture, and they also tended to lose Foxp3 following the repeated stimulation. Finally, a proportion of expanded HY-specific Tregs secreted IL2 in response to their cognate peptide, and this finding could be confirmed using Tregs from Foxp3GFP reporter mice. We not only demonstrated that expanded Tregs are superior to fresh Tregs in suppressing T cell responses against alloantigens, but also revealed some novel immunobiological properties of expended Tregs which are very instructive for modifying current Treg expansion procedures.


Blood | 2009

C1q enhances IFN-γ production by antigen-specific T cells via the CD40 costimulatory pathway on dendritic cells

Paramita Baruah; Ingrid E. Dumitriu; Talat H. Malik; H. Terence Cook; Julian Dyson; Diane Scott; Elizabeth Simpson; Marina Botto

Dendritic cells (DCs) are known to produce C1q, the initiator of the classical complement pathway. We demonstrate that murine DCs deficient in C1q (C1qa(-/-)) are poorer than wild-type (WT) DCs at eliciting the proliferation and Th1 differentiation of antigen-specific T cells. These defects result from decreased production of IL-12p70 by C1qa(-/-) DCs and impaired expression of costimulatory molecules CD80 and CD86 in response to CD40 ligation. The defective production of IL-12p70 and the reduced expression of CD80 and CD86 by C1qa(-/-) DCs were specifically mediated via CD40 ligation, as normal levels of IL-12p70 and CD80/86 were observed after ligation of Toll-like receptors (TLRs) on C1qa(-/-) DCs. CD40 ligation on C1qa(-/-) DCs, but not TLR ligation, results in decreased phosphorylation of p38 and ERK1/2 kinases. A strong colocalization of CD40 and C1q was observed by confocal microscopy upon CD40 ligation (but not TLR ligation) on DCs. Furthermore, human DCs from 2 C1q-deficient patients were found to have impaired IL-12p70 production in response to CD40L stimulation. Our novel data suggest that C1q augments the production of IL-12p70 by mouse and human DCs after CD40 triggering and plays important roles in sustaining the maturation of DCs and guiding the activation of T cells.


Journal of Immunology | 2006

Role of Immunoproteasomes in Cross-Presentation

Michael J. Palmowski; Uzi Gileadi; Mariolina Salio; Awen Myfanwy Gallimore; Maggie Millrain; Edward James; Caroline Addey; Diane Scott; Julian Dyson; Elizabeth Simpson; Vincenzo Cerundolo

The evidence that proteasomes are involved in the processing of cross-presented proteins is indirect and based on the in vitro use of proteasome inhibitors. It remains, therefore, unclear whether cross-presentation of MHC class I peptide epitopes can occur entirely within phagolysosomes or whether it requires proteasome degradation. To address this question, we studied in vivo cross-presentation of an immunoproteasome-dependent epitope. First, we demonstrated that generation of the immunodominant HY Uty246–254 epitope is LMP7 dependent, resulting in the lack of rejection of male LMP7-deficient (LMP7−/−) skin grafts by female LMP7−/− mice. Second, we ruled out an altered Uty246–254-specific T cell repertoire in LMP7−/− female mice and demonstrated efficient Uty246–254 presentation by re-expressing LMP7 in male LMP7−/− cells. Finally, we observed that LMP7 expression significantly enhanced cross-priming of Uty246–254-specific T cells in vivo. The observations that male skin grafts are not rejected by LMP7−/− female mice and that presentation of a proteasome-dependent peptide is not efficiently rescued by alternative cross-presentation pathways provide strong evidence that proteasomes play an important role in cross-priming events.


Journal of Immunology | 2000

Activated Murine Endothelial Cells Have Reduced Immunogenicity for CD8+ T Cells: A Mechanism of Immunoregulation?

Federica M. Marelli-Berg; Diane Scott; Istvan Bartok; Emma Peek; Julian Dyson; Robert I. Lechler

The immunogenic properties of primary cultures of murine lung microvascular endothelial cells (EC) were analyzed. Resting endothelial cells were found to constitutively express low levels of MHC class I and CD80 molecules. IFN-γ treatment of EC resulted in a marked up-regulation of MHC class I, but no change was observed in the level of CD80 expression. No CD86 molecules were detectable under either condition. The ability of peptide-pulsed EC to induce the proliferation of either the HY-specific, H2-Kk-restricted CD8+ T cell clone (C6) or C6 TCR-transgenic naive CD8+ T cells was analyzed. Resting T cells were stimulated to divide by quiescent peptide-prepulsed EC, while peptide-pulsed, cytokine-activated EC lost the ability to induce T cell division. Furthermore, Ag presentation by cytokine-activated EC induced CD8+ T cell hyporesponsiveness. The immunogenicity of activated EC could be restored by adding nonsaturating concentrations of anti-H2-Kk Ab in the presence of an optimal concentration of cognate peptide. This is consistent with the suggestion that the ratio of TCR engagement to costimulation determines the outcome of T cell recognition. In contrast, activated peptide-pulsed EC were killed more efficiently by fully differentiated effector CD8+ T cells. Finally, evidence is provided that Ag recognition of EC can profoundly affect the transendothelial migration of CD8+ T cells. Taken together, these results suggest that EC immunogenicity is regulated in a manner that contributes to peripheral tolerance.


Journal of Molecular Medicine | 1997

Why do some females reject males? The molecular basis for male-specific graft rejection

Diane Scott; I. E. Ehrmann; Pamela Ellis; Phillip Chandler; Elizabeth Simpson

Abstract The male-specific minor histocompatibility antigen H-Y plays an important role in both graft rejection and graft-versus-host disease following transplantation of male tissue into females that are completely matched at the major histocompatibility loci. The recent identification of two peptides that, in association with the mouse H-2Kk or human HLA B7 major histocompatibility class I molecules, are recognised by H-Y-specific T cells, has provided evidence for the molecular basis for such anti-H-Y responses. These peptides are encoded by the mouse and human homologues of a ubiquitously expressed Y chromosome gene, Smcy, whilst the equivalent peptides encoded by the X chromosome homologues of this gene fail to be recognised. Genetic studies have demonstrated that, as is the case for other minor histocompatibility antigens, peptide epitopes from several closely linked genes may be required to interact in order to elicit a response against H-Y. Definition of the peptides and the genes that encode these epitopes will allow the devopment of tolerogenic protocols that could specifically down-modulate the response to H-Y and perhaps even other minor histocompatibility antigens.

Collaboration


Dive into the Diane Scott's collaboration.

Top Co-Authors

Avatar

Julian Dyson

Imperial College London

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Simpson

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Simpson

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Federica M. Marelli-Berg

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Marina Botto

Imperial College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge