Network


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

Hotspot


Dive into the research topics where Lauren M. Webb is active.

Publication


Featured researches published by Lauren M. Webb.


The EMBO Journal | 2014

RNA:DNA hybrids are a novel molecular pattern sensed by TLR9.

Rachel E. Rigby; Lauren M. Webb; Karen J. Mackenzie; Yue Li; Andrea Leitch; Martin A. M. Reijns; Rachel J. Lundie; Ailsa Revuelta; Donald J. Davidson; Sandra S. Diebold; Yorgo Modis; Andrew S. MacDonald; Andrew P. Jackson

The sensing of nucleic acids by receptors of the innate immune system is a key component of antimicrobial immunity. RNA:DNA hybrids, as essential intracellular replication intermediates generated during infection, could therefore represent a class of previously uncharacterised pathogen‐associated molecular patterns sensed by pattern recognition receptors. Here we establish that RNA:DNA hybrids containing viral‐derived sequences efficiently induce pro‐inflammatory cytokine and antiviral type I interferon production in dendritic cells. We demonstrate that MyD88‐dependent signalling is essential for this cytokine response and identify TLR9 as a specific sensor of RNA:DNA hybrids. Hybrids therefore represent a novel molecular pattern sensed by the innate immune system and so could play an important role in host response to viruses and the pathogenesis of autoimmune disease.


Nature Communications | 2015

A dominant role for the methyl-CpG-binding protein Mbd2 in controlling Th2 induction by dendritic cells

Peter C. Cook; Heather Owen; Aimée M. Deaton; Jessica G. Borger; Sheila Brown; Thomas Clouaire; Gareth-Rhys Jones; Lucy H. Jones; Rachel J. Lundie; Angela K. Marley; Vicky L. Morrison; Alexander T. Phythian-Adams; Elisabeth Wachter; Lauren M. Webb; Tara E. Sutherland; Graham D. Thomas; John R. Grainger; Jim Selfridge; Andrew N. J. McKenzie; Judith E. Allen; Susanna C. Fagerholm; Rick M. Maizels; Alasdair Ivens; Adrian Bird; Andrew S. MacDonald

Dendritic cells (DCs) direct CD4+ T-cell differentiation into diverse helper (Th) subsets that are required for protection against varied infections. However, the mechanisms used by DCs to promote Th2 responses, which are important both for immunity to helminth infection and in allergic disease, are currently poorly understood. We demonstrate a key role for the protein methyl-CpG-binding domain-2 (Mbd2), which links DNA methylation to repressive chromatin structure, in regulating expression of a range of genes that are associated with optimal DC activation and function. In the absence of Mbd2, DCs display reduced phenotypic activation and a markedly impaired capacity to initiate Th2 immunity against helminths or allergens. These data identify an epigenetic mechanism that is central to the activation of CD4+ T-cell responses by DCs, particularly in Th2 settings, and reveal methyl-CpG-binding proteins and the genes under their control as possible therapeutic targets for type-2 inflammation.


Journal of Immunology | 2014

MyD88 Signaling Inhibits Protective Immunity to the Gastrointestinal Helminth Parasite Heligmosomoides polygyrus

Lisa A. Reynolds; Yvonne Harcus; Katherine A. Smith; Lauren M. Webb; James P. Hewitson; Ewan A. Ross; Sheila Brown; Satoshi Uematsu; Shizuo Akira; David Gray; Mohini Gray; Andrew S. MacDonald; Adam F. Cunningham; Rick M. Maizels

Helminth parasites remain one of the most common causes of infections worldwide, yet little is still known about the immune signaling pathways that control their expulsion. C57BL/6 mice are chronically susceptible to infection with the gastrointestinal helminth parasite Heligmosomoides polygyrus. In this article, we report that C57BL/6 mice lacking the adapter protein MyD88, which mediates signaling by TLRs and IL-1 family members, showed enhanced immunity to H. polygyrus infection. Alongside increased parasite expulsion, MyD88-deficient mice showed heightened IL-4 and IL-17A production from mesenteric lymph node CD4+ cells. In addition, MyD88−/− mice developed substantial numbers of intestinal granulomas around the site of infection, which were not seen in MyD88-sufficient C57BL/6 mice, nor when signaling through the adapter protein TRIF (TIR domain–containing adapter–inducing IFN-β adapter protein) was also ablated. Mice deficient solely in TLR2, TLR4, TLR5, or TLR9 did not show enhanced parasite expulsion, suggesting that these TLRs signal redundantly to maintain H. polygyrus susceptibility in wild-type mice. To further investigate signaling pathways that are MyD88 dependent, we infected IL-1R1−/− mice with H. polygyrus. This genotype displayed heightened granuloma numbers compared with wild-type mice, but without increased parasite expulsion. Thus, the IL-1R–MyD88 pathway is implicated in inhibiting granuloma formation; however, protective immunity in MyD88-deficient mice appears to be granuloma independent. Like IL-1R1−/− and MyD88−/− mice, animals lacking signaling through the type 1 IFN receptor (i.e., IFNAR1−/−) also developed intestinal granulomas. Hence, IL-1R1, MyD88, and type 1 IFN receptor signaling may provide pathways to impede granuloma formation in vivo, but additional MyD88-mediated signals are associated with inhibition of protective immunity in susceptible C57BL/6 mice.


Journal of Immunology | 2012

Concurrent bacterial stimulation alters the function of helminth-activated dendritic cells, resulting in IL-17 induction.

Georgia Perona-Wright; Rachel J. Lundie; Stephen J. Jenkins; Lauren M. Webb; Richard K. Grencis; Andrew S. MacDonald

Infection with schistosome helminths is associated with granulomatous inflammation that forms around parasite eggs trapped in host tissues. In severe cases, the resulting fibrosis can lead to organ failure, portal hypertension, and fatal bleeding. Murine studies identified IL-17 as a critical mediator of this immunopathology, and mouse strains that produce high levels of IL-17 in response to schistosome infection show increased mortality. In this article, we demonstrate that schistosome-specific IL-17 induction by dendritic cells from low-pathology C57BL/6 mice is normally regulated by their concomitant induction of IL-10. Simultaneous stimulation of schistosome-exposed C57BL/6 dendritic cells with a heat-killed bacterium enabled these cells to overcome IL-10 regulation and induce IL-17, even in wild-type C57BL/6 recipients. This schistosome-specific IL-17 was dependent on IL-6 production by the copulsed dendritic cells. Coimmunization of C57BL/6 animals with bacterial and schistosome Ags also resulted in schistosome-specific IL-17, and this response was enhanced in the absence of IL-10–mediated immune regulation. Together, our data suggest that the balance of pro- and anti-inflammatory cytokines that determines the severity of pathology during schistosome infection can be influenced not only by host and parasite, but also by concurrent bacterial stimulation.


PLOS Pathogens | 2014

The Secreted Triose Phosphate Isomerase of Brugia malayi Is Required to Sustain Microfilaria Production In Vivo

James P. Hewitson; Dominik Rückerl; Yvonne Harcus; Janice Murray; Lauren M. Webb; Simon A. Babayan; Judith E. Allen; Agnes Kurniawan; Rick M. Maizels

Human lymphatic filariasis is a major tropical disease transmitted through mosquito vectors which take up microfilarial larvae from the blood of infected subjects. Microfilariae are produced by long-lived adult parasites, which also release a suite of excretory-secretory products that have recently been subject to in-depth proteomic analysis. Surprisingly, the most abundant secreted protein of adult Brugia malayi is triose phosphate isomerase (TPI), a glycolytic enzyme usually associated with the cytosol. We now show that while TPI is a prominent target of the antibody response to infection, there is little antibody-mediated inhibition of catalytic activity by polyclonal sera. We generated a panel of twenty-three anti-TPI monoclonal antibodies and found only two were able to block TPI enzymatic activity. Immunisation of jirds with B. malayi TPI, or mice with the homologous protein from the rodent filaria Litomosoides sigmodontis, failed to induce neutralising antibodies or protective immunity. In contrast, passive transfer of neutralising monoclonal antibody to mice prior to implantation with adult B. malayi resulted in 60–70% reductions in microfilarial levels in vivo and both oocyte and microfilarial production by individual adult females. The loss of fecundity was accompanied by reduced IFNγ expression by CD4+ T cells and a higher proportion of macrophages at the site of infection. Thus, enzymatically active TPI plays an important role in the transmission cycle of B. malayi filarial parasites and is identified as a potential target for immunological and pharmacological intervention against filarial infections.


Immunology and Cell Biology | 2016

A central role for hepatic conventional dendritic cells in supporting Th2 responses during helminth infection

Rachel J. Lundie; Lauren M. Webb; Angela K. Marley; Alexander T. Phythian-Adams; Peter C. Cook; Lucy H. Jackson-Jones; Sheila Brown; Rick M. Maizels; Louis Boon; Meredith O'Keeffe; Andrew S. MacDonald

Dendritic cells (DCs) are the key initiators of T‐helper (Th) 2 immune responses against the parasitic helminth Schistosoma mansoni. Although the liver is one of the main sites of antigen deposition during infection with this parasite, it is not yet clear how distinct DC subtypes in this tissue respond to S. mansoni antigens in vivo, or how the liver microenvironment might influence DC function during establishment of the Th2 response. In this study, we show that hepatic DC subsets undergo distinct activation processes in vivo following murine infection with S. mansoni. Conventional DCs (cDCs) from schistosome‐infected mice upregulated expression of the costimulatory molecule CD40 and were capable of priming naive CD4+ T cells, whereas plasmacytoid DCs (pDCs) upregulated expression of MHC class II, CD86 and CD40 but were unable to support the expansion of either naive or effector/memory CD4+ T cells. Importantly, in vivo depletion of pDCs revealed that this subset was dispensable for either maintenance or regulation of the hepatic Th2 effector response during acute S. mansoni infection. Our data provides strong evidence that S. mansoni infection favors the establishment of an immunogenic, rather than tolerogenic, liver microenvironment that conditions cDCs to initiate and maintain Th2 immunity in the context of ongoing antigen exposure.


The Journal of Allergy and Clinical Immunology | 2017

Tumor progression locus 2 reduces severe allergic airway inflammation by inhibiting Ccl24 production in dendritic cells.

Yashaswini Kannan; Yanda Li; Stephanie M. Coomes; Isobel S. Okoye; Victoria S. Pelly; Srividya Sriskantharajah; Eva Gückel; Lauren M. Webb; Stephanie Czieso; Nikolay Nikolov; Andrew S. MacDonald; Steven C. Ley; Mark S. Wilson

Background: The molecular and cellular pathways driving the pathogenesis of severe asthma are poorly defined. Tumor progression locus 2 (TPL‐2) (COT, MAP3K8) kinase activates the MEK1/2‐extracellular‐signal regulated kinase 1/2 MAP kinase signaling pathway following Toll‐like receptor, TNFR1, and IL‐1R stimulation. Objective: TPL‐2 has been widely described as a critical regulator of inflammation, and we sought to investigate the role of TPL‐2 in house dust mite (HDM)‐mediated allergic airway inflammation. Methods: A comparative analysis of wild‐type and Map3k8−/− mice was conducted. Mixed bone marrow chimeras, conditional knockout mice, and adoptive transfer models were also used. Differential cell counts were performed on the bronchoalveolar lavage fluid, followed by histological analysis of lung sections. Flow cytometry and quantitative PCR was used to measure type 2 cytokines. ELISA was used to assess the production of IgE, type 2 cytokines, and Ccl24. RNA sequencing was used to characterize dendritic cell (DC) transcripts. Results: TPL‐2 deficiency led to exacerbated HDM‐induced airway allergy, with increased airway and tissue eosinophilia, lung inflammation, and IL‐4, IL‐5, IL‐13, and IgE production. Increased airway allergic responses in Map3k8−/− mice were not due to a cell‐intrinsic role for TPL‐2 in T cells, B cells, or LysM+ cells but due to a regulatory role for TPL‐2 in DCs. TPL‐2 inhibited Ccl24 expression in lung DCs, and blockade of Ccl24 prevented the exaggerated airway eosinophilia and lung inflammation in mice given HDM‐pulsed Map3k8−/− DCs. Conclusions: TPL‐2 regulates DC‐derived Ccl24 production to prevent severe type 2 airway allergy in mice.


The EMBO Journal | 2017

Type I interferon is required for T helper (Th) 2 induction by dendritic cells

Lauren M. Webb; Rachel J. Lundie; Jessica G. Borger; Sheila Brown; Lisa M. Connor; Adam N.R. Cartwright; Annette M. Dougall; Ruud H. P. Wilbers; Peter C. Cook; Lucy H. Jackson-Jones; Alexander T. Phythian-Adams; Cecilia Johansson; Daniel M. Davis; Benjamin G Dewals; Franca Ronchese; Andrew S. MacDonald

Type 2 inflammation is a defining feature of infection with parasitic worms (helminths), as well as being responsible for widespread suffering in allergies. However, the precise mechanisms involved in T helper (Th) 2 polarization by dendritic cells (DCs) are currently unclear. We have identified a previously unrecognized role for type I IFN (IFN‐I) in enabling this process. An IFN‐I signature was evident in DCs responding to the helminth Schistosoma mansoni or the allergen house dust mite (HDM). Further, IFN‐I signaling was required for optimal DC phenotypic activation in response to helminth antigen (Ag), and efficient migration to, and localization with, T cells in the draining lymph node (dLN). Importantly, DCs generated from Ifnar1−/− mice were incapable of initiating Th2 responses in vivo. These data demonstrate for the first time that the influence of IFN‐I is not limited to antiviral or bacterial settings but also has a central role to play in DC initiation of Th2 responses.


The EMBO Journal | 2017

A central role for Type I IFN in Th2 response induction by dendritic cells

Lauren M. Webb; Rachel J. Lundie; Jessica G. Borger; Sheila Brown; Lisa C. Connor; Adam N.R. Cartwright; Annette M. Dougall; Ruud H. P. Wilbers; Peter C. Cook; Lucy H. Jackson-Jones; Alexander T. Phytian-Adams; Cecilia Johansson; Daniel M. Davis; Benjamin G Dewals; Franca Ronchese; Andrew S. MacDonald


Immunology | 2012

Epigenetic control of Th2 induction by dendritic cells

Peter C. Cook; Aimée M. Deaton; Heather Owen; Graham D. Thomas; Lucy H. Jones; Alex T. Phythian-Adams; Rachel J. Lundie; Lauren M. Webb; Angela K. Marley; Jessica G. Borger; Tara E. Sutherland; John Allen; John Grainger; Rick M. Maizels; Alasdair Ivens; Adrian Bird; Andrew S. MacDonald

Collaboration


Dive into the Lauren M. Webb's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter C. Cook

University of Manchester

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sheila Brown

University of Edinburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge