Network


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

Hotspot


Dive into the research topics where Rebecca Leyland is active.

Publication


Featured researches published by Rebecca Leyland.


Immunological Reviews | 2013

miR‐155: an ancient regulator of the immune system

Elena Vigorito; Susan Kohlhaas; Dong Lu; Rebecca Leyland

MicroRNAs (miRNAs) are a newly recognized class of regulatory genes which repress the expression of protein‐coding genes. Numerous studies have uncovered a complex role for miRNAs regulating many aspects of a variety of cellular processes including cell growth, differentiation, and lineage commitment. In the immune system, miR‐155 is unique in its ability to shape the transcriptome of activated myeloid and lymphoid cells controlling diverse biological functions ranging from inflammation to immunological memory. Not surprisingly, a tight control of miR‐155 expression is required to avoid malignant transformation, as evidenced by miR‐155 overexpression in many cancers of B‐cell origin. In this review, we discuss the potential of miR‐155 as a molecular target for therapeutic intervention and discuss the function of miR‐155 in the context of protective immunity. We first look back into the emergence of miR‐155 in evolution, which is coincidental with the emergence of the ancestors of the antigen receptors. We then summarize what we have learned about the role of miR‐155 in the regulation of lymphoid subsets at the cellular and molecular level in the context of recent progress in this field.


Molecular and Cellular Biology | 2012

MicroRNA-155 Is Required for Mycobacterium bovis BCG-Mediated Apoptosis of Macrophages

Devram Sampat Ghorpade; Rebecca Leyland; Mariola Kurowska-Stolarska; Shripad A. Patil; Kithiganahalli Narayanaswamy Balaji

ABSTRACT Pathogenic mycobacteria, including Mycobacterium tuberculosis and Mycobacterium bovis, cause significant morbidity and mortality worldwide. However, the vaccine strain Mycobacterium bovis BCG, unlike virulent strains, triggers extensive apoptosis of infected macrophages, a step necessary for the elicitation of robust protective immunity. We here demonstrate that M. bovis BCG triggers Toll-like receptor 2 (TLR2)-dependent microRNA-155 (miR-155) expression, which involves signaling cross talk among phosphatidylinositol 3-kinase (PI3K), protein kinase Cδ (PKCδ), and mitogen-activated protein kinases (MAPKs) and recruitment of NF-κB and c-ETS to miR-155 promoter. Genetic and signaling perturbations presented the evidence that miR-155 regulates PKA signaling by directly targeting a negative regulator of PKA, protein kinase inhibitor alpha (PKI-α). Enhanced activation of PKA signaling resulted in the generation of PKA C-α; phosphorylation of MSK1, cyclic AMP response element binding protein (CREB), and histone H3; and recruitment of phospho-CREB to the apoptotic gene promoters. The miR-155-triggered activation of caspase-3, BAK1, and cytochrome c translocation involved signaling integration of MAPKs and epigenetic or posttranslational modification of histones or CREB. Importantly, M. bovis BCG infection-induced apoptosis was severely compromised in macrophages derived from miR-155 knockout mice. Gain-of-function and loss-of-function studies validated the requirement of miR-155 for M. bovis BCGs ability to trigger apoptosis. Overall, M. bovis BCG-driven miR-155 dictates cell fate decisions of infected macrophages, strongly implicating a novel role for miR-155 in orchestrating cellular reprogramming during immune responses to mycobacterial infection.


Journal of Neurochemistry | 2009

Characterisation of 5‐HT3C, 5‐HT3D and 5‐HT3E receptor subunits: evolution, distribution and function

Joanna D. Holbrook; Catherine H. Gill; Noureddine Zebda; Jon P. Spencer; Rebecca Leyland; Kim Rance; Han Trinh; Gemma Balmer; Fiona M. Kelly; Shahnaz P. Yusaf; Nicola Courtenay; Jane Luck; Andrew Rhodes; Sundip Modha; Stephen Moore; Gareth J. Sanger; Martin J. Gunthorpe

The 5‐HT3 receptor is a member of the ‘Cys‐loop’ family of ligand‐gated ion channels that mediate fast excitatory and inhibitory transmission in the nervous system. Current evidence points towards native 5‐HT3 receptors originating from homomeric assemblies of 5‐HT3A or heteromeric assembly of 5‐HT3A and 5‐HT3B. Novel genes encoding 5‐HT3C, 5‐HT3D, and 5‐HT3E have recently been described but the functional importance of these proteins is unknown. In the present study, in silico analysis (confirmed by partial cloning) indicated that 5‐HT3C, 5‐HT3D, and 5‐HT3E are not human–specific as previously reported: they are conserved in multiple mammalian species but are absent in rodents. Expression profiles of the novel human genes indicated high levels in the gastrointestinal tract but also in the brain, Dorsal Root Ganglion (DRG) and other tissues. Following the demonstration that these subunits are expressed at the cell membrane, the functional properties of the recombinant human subunits were investigated using patch clamp electrophysiology. 5‐HT3C, 5‐HT3D, and 5‐HT3E were all non‐functional when expressed alone. Co‐transfection studies to determine potential novel heteromeric receptor interactions with 5‐HT3A demonstrated that the expression or function of the receptor was modified by 5‐HT3C and 5‐HT3E, but not 5‐HT3D. The lack of distinct effects on current rectification, kinetics or pharmacology of 5‐HT3A receptors does not however provide unequivocal evidence to support a direct contribution of 5‐HT3C or 5‐HT3E to the lining of the ion channel pore of novel heteromeric receptors. The functional and pharmacological contributions of these novel subunits to human biology and diseases such as irritable bowel syndrome for which 5‐HT3 receptor antagonists have major clinical usage, therefore remains to be fully determined.


Journal of Experimental Medicine | 2014

The miR-155–PU.1 axis acts on Pax5 to enable efficient terminal B cell differentiation

Dong Lu; Rinako Nakagawa; Sandra Lazzaro; Philipp Staudacher; Cei Abreu-Goodger; Tom Henley; Sara Boiani; Rebecca Leyland; Alison Galloway; Simon Andrews; Geoffrey W. Butcher; Stephen L. Nutt; Martin Turner; Elena Vigorito

Lu et al. disrupt the interaction between miR-155 and the transcription factor PU.1 by specifically removing miR-155–binding site from PU.1 mRNA in mice. They show that this interaction is required for plasma cell formation and extrafollicular response to immunization in vivo.


Cancer immunology research | 2017

Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery

Suzanne Mosely; John E. Prime; Richard Sainson; Jens-Oliver Koopmann; Dennis Wang; Danielle Greenawalt; Miika Ahdesmaki; Rebecca Leyland; Stefanie Mullins; Luciano Pacelli; Danielle Marcus; Judith Anderton; Amanda Watkins; Jane Coates Ulrichsen; Philip Brohawn; Brandon W. Higgs; Matthew McCourt; Hazel Jones; James Harper; Michelle Morrow; Viia Valge-Archer; Ross Stewart; Simon J. Dovedi; Robert W. Wilkinson

Murine syngeneic tumor models are used to study responses to antitumor immunotherapies. To rationalize model selection, the underlying genetic and immunologic biology of the models was analyzed, allowing parallels to be drawn between models and human disease phenotypes. Murine syngeneic tumor models are critical to novel immuno-based therapy development, but the molecular and immunologic features of these models are still not clearly defined. The translational relevance of differences between the models is not fully understood, impeding appropriate preclinical model selection for target validation, and ultimately hindering drug development. Across a panel of commonly used murine syngeneic tumor models, we showed variable responsiveness to immunotherapies. We used array comparative genomic hybridization, whole-exome sequencing, exon microarray analysis, and flow cytometry to extensively characterize these models, which revealed striking differences that may underlie these contrasting response profiles. We identified strong differential gene expression in immune-related pathways and changes in immune cell–specific genes that suggested differences in tumor immune infiltrates between models. Further investigation using flow cytometry showed differences in both the composition and magnitude of the tumor immune infiltrates, identifying models that harbor “inflamed” and “non-inflamed” tumor immune infiltrate phenotypes. We also found that immunosuppressive cell types predominated in syngeneic mouse tumor models that did not respond to immune-checkpoint blockade, whereas cytotoxic effector immune cells were enriched in responsive models. A cytotoxic cell–rich tumor immune infiltrate has been correlated with increased efficacy of immunotherapies in the clinic, and these differences could underlie the varying response profiles to immunotherapy between the syngeneic models. This characterization highlighted the importance of extensive profiling and will enable investigators to select appropriate models to interrogate the activity of immunotherapies as well as combinations with targeted therapies in vivo. Cancer Immunol Res; 5(1); 29–41. ©2016 AACR.


Journal of Clinical Investigation | 2016

MicroRNA-155 controls affinity-based selection by protecting c-MYC+ B cells from apoptosis

Rinako Nakagawa; Rebecca Leyland; Michael Meyer-Hermann; Dong Lu; Martin Turner; Giuseppina Arbore; Tri Giang Phan; Robert Brink; Elena Vigorito

The production of high-affinity antibodies by B cells is essential for pathogen clearance. Antibody affinity for antigen is increased through the affinity maturation in germinal centers (GCs). This is an iterative process in which B cells cycle between proliferation coupled with the acquisition of mutations and antigen-based positive selection, resulting in retention of the highest-affinity B cell clones. The posttranscriptional regulator microRNA-155 (miR-155) is critical for efficient affinity maturation and the maintenance of the GCs; however, the cellular and molecular mechanism by which miR-155 regulates GC responses is not well understood. Here, we utilized a miR-155 reporter mouse strain and showed that miR-155 is coexpressed with the proto-oncogene encoding c-MYC in positively selected B cells. Functionally, miR-155 protected positively selected c-MYC+ B cells from apoptosis, allowing clonal expansion of this population, providing an explanation as to why Mir155 deletion impairs affinity maturation and promotes the premature collapse of GCs. We determined that miR-155 directly inhibits the Jumonji family member JARID2, which enhances B cell apoptosis when overexpressed, and thereby promotes GC B cell survival. Our findings also suggest that there is cooperation between c-MYC and miR-155 during the normal GC response, a cooperation that may explain how c-MYC and miR-155 can collaboratively function as oncogenes.


Clinical Cancer Research | 2017

A Novel Murine GITR Ligand Fusion Protein Induces Antitumor Activity as a Monotherapy That Is Further Enhanced in Combination with an OX40 Agonist

Rebecca Leyland; Amanda Watkins; Kathy Mulgrew; Nicholas Holoweckyj; Lisa Bamber; Natalie Tigue; Emily Offer; John Andrews; Li Yan; Stefanie Mullins; Michael Oberst; Jane Coates Ulrichsen; David A Leinster; Kelly McGlinchey; Lesley Young; Michelle Morrow; Scott A. Hammond; Philip R Mallinder; Athula Herath; Ching Ching Leow; Robert W. Wilkinson; Ross Stewart

Purpose: To generate and characterize a murine GITR ligand fusion protein (mGITRL-FP) designed to maximize valency and the potential to agonize the GITR receptor for cancer immunotherapy. Experimental Design: The EC50 value of the mGITRL-FP was compared with an anti-GITR antibody in an in vitro agonistic cell–based reporter assay. We assessed the impact of dose, schedule, and Fc isotype on antitumor activity and T-cell modulation in the CT26 tumor model. The activity of the mGITRL-FP was compared with an agonistic murine OX40L-FP targeting OX40, in CT26 and B16F10-Luc2 tumor models. Combination of the mGITRL-FP with antibodies targeting PD-L1, PD-1, or CTLA-4 was analyzed in mice bearing CT26 tumors. Results: The mGITRL-FP had an almost 50-fold higher EC50 value compared with an anti-murine GITR antibody. Treatment of CT26 tumor-bearing mice with mGITRL-FP–mediated significant antitumor activity that was dependent on isotype, dose, and duration of exposure. The antitumor activity could be correlated with the increased proliferation of peripheral CD8+ and CD4+ T cells and a significant decrease in the frequency of intratumoral Tregs. The combination of mGITRL-FP with mOX40L-FP or checkpoint inhibitor antagonists enhanced antitumor immunity above that of monotherapy treatment. Conclusions: These results suggest that therapeutically targeting GITR represents a unique approach to cancer immunotherapy and suggests that a multimeric fusion protein may provide increased agonistic potential versus an antibody. In addition, these data provide, for the first time, early proof of concept for the potential combination of GITR targeting agents with OX40 agonists and PD-L1 antagonists. Clin Cancer Res; 23(13); 3416–27. ©2017 AACR.


Oncotarget | 2016

Phenotypic screening reveals TNFR2 as a promising target for cancer immunotherapy

Geoffrey S. Williams; Bina Mistry; Sandrine Guillard; Jane Coates Ulrichsen; Alan Sandercock; Jun Wang; Andrea González-Muñoz; Julie Parmentier; Chelsea Black; Jo Soden; Jim Freeth; Jelena Jovanović; Rebecca Leyland; Rafia S. Al-Lamki; Andrew J. Leishman; Steven J. Rust; Ross Stewart; Lutz Jermutus; John R. Bradley; Vahe Bedian; Viia Valge-Archer; Ralph Minter; Robert W. Wilkinson

Antibodies that target cell-surface molecules on T cells can enhance anti-tumor immune responses, resulting in sustained immune-mediated control of cancer. We set out to find new cancer immunotherapy targets by phenotypic screening on human regulatory T (Treg) cells and report the discovery of novel activators of tumor necrosis factor receptor 2 (TNFR2) and a potential role for this target in immunotherapy. A diverse phage display library was screened to find antibody mimetics with preferential binding to Treg cells, the most Treg-selective of which were all, without exception, found to bind specifically to TNFR2. A subset of these TNFR2 binders were found to agonise the receptor, inducing iκ-B degradation and NF-κB pathway signalling in vitro. TNFR2 was found to be expressed by tumor-infiltrating Treg cells, and to a lesser extent Teff cells, from three lung cancer patients, and a similar pattern was also observed in mice implanted with CT26 syngeneic tumors. In such animals, TNFR2-specific agonists inhibited tumor growth, enhanced tumor infiltration by CD8+ T cells and increased CD8+ T cell IFN-γ synthesis. Together, these data indicate a novel mechanism for TNF-α-independent TNFR2 agonism in cancer immunotherapy, and demonstrate the utility of target-agnostic screening in highlighting important targets during drug discovery.


Cancer Research | 2016

Abstract 561: MEDI1873: A novel hexameric GITRL fusion protein with potent agonsitic and immunomodulatory activities in preclinical systems

Ross Stewart; Natalie Tigue; Samantha Ireland; James Hair; Lisa Bamber; Michael Oberst; Rebecca Leyland; Amanda Watkins; Maureen Kennedy; Cann Jennifer; Lesley Young; Robert W. Wilkinson

Glucocorticoid-induced TNFR-related protein (GITR) is a member of the tumor necrosis factor receptor (TNFR) superfamily. GITR is expressed constitutively on regulatory T cells (Tregs) and is up-regulated on other T cells following activation. Agonistic antibodies to GITR have demonstrated significant activity in preclinical models of cancer. Here we describe the generation and characterisation of a GITR ligand (GITRL) fusion protein (FP) (MEDI1873), currently in phase 1 clinical trials. Protein engineering was used to generate a series of GITRL FPs, which were screened using a high throughput reporter gene assay for GITR signalling. The most potent fusion protein resulted in a 20 times greater maximal signal and a 5 times higher EC50 when compared to a GITR targeting antibody. This increased potency was considered to be a result of the enhanced valency achieved by the hexameric format. Two versions of GITRL FP, MEDI1873 and MEDI5607, bearing an IgG1 and IgG4 Fc respectively, both demonstrated equivalent potency in a reporter assay and were able to enhance T-cell activation, with respect to proliferation and cytokine release, and to overcome the suppressive effect of Tregs, in primary human cell based assays. Assessment of two surrogate mouse GITRL FPs in the CT26 model of colorectal cancer indicated that the version with increased binding to Fc gamma receptors resulted in increased activity, coincident with an increased depletion of intratumoral Tregs, likely through Fc mediated effector functions. A comparison of GITR expression on Tregs and effector T cells in mouse and human, via flow cytometry, indicated a similar pattern of expression across species, with significantly higher expression observed on Tregs. Immunohistochemical analysis indicated the presence of high levels of both GITR and FoxP3 in sections from human tumors; suggesting that the intratumoral Treg depletion observed in mice could also occur in humans. Both MEDI1873 and MEDI5607 demonstrated enhanced binding to Fc gamma receptors when compared to antibody controls of the same isotype, again considered to be a result of their increased valency. However, only MEDI1873 was able to mediate ADCC against activated T cells in vitro; resulting in an increase in the CD8:CD4 ratio within the culture. As a result of these studies, MEDI1873 was selected as an optimal GITR targeting agent that possessed the ability to both agonise GITR and to modulate Tregs through suppression and/or depletion. MEDI1873 is currently being assessed in a phase 1 clinical study (NCT02583165) in patients with solid tumors.


Frontiers in Immunology | 2018

Epigenomic Modifications Mediating Antibody Maturation

Emily C. Sheppard; Rikke Brandstrup Morrish; Michael J. Dillon; Rebecca Leyland; Richard Chahwan

Epigenetic modifications, such as histone modifications, DNA methylation status, and non-coding RNAs (ncRNA), all contribute to antibody maturation during somatic hypermutation (SHM) and class-switch recombination (CSR). Histone modifications alter the chromatin landscape and, together with DNA primary and tertiary structures, they help recruit Activation-Induced Cytidine Deaminase (AID) to the immunoglobulin (Ig) locus. AID is a potent DNA mutator, which catalyzes cytosine-to-uracil deamination on single-stranded DNA to create U:G mismatches. It has been shown that alternate chromatin modifications, in concert with ncRNAs and potentially DNA methylation, regulate AID recruitment and stabilize DNA repair factors. We, hereby, assess the combination of these distinct modifications and discuss how they contribute to initiating differential DNA repair pathways at the Ig locus, which ultimately leads to enhanced antibody–antigen binding affinity (SHM) or antibody isotype switching (CSR). We will also highlight how misregulation of epigenomic regulation during DNA repair can compromise antibody development and lead to a number of immunological syndromes and cancer.

Collaboration


Dive into the Rebecca Leyland'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

Dong Lu

Wellcome Trust Sanger Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge