11 research outputs found

    Impaired expression of metallothioneins contributes to allergen-induced inflammation in patients with atopic dermatitis

    Get PDF
    Regulation of cutaneous immunity is severely compromised in inflammatory skin disease. To investigate the molecular crosstalk underpinning tolerance versus inflammation in atopic dermatitis, we utilise a human in vivo allergen challenge study, exposing atopic dermatitis patients to house dust mite. Here we analyse transcriptional programmes at the population and single cell levels in parallel with immunophenotyping of cutaneous immunocytes revealed a distinct dichotomy in atopic dermatitis patient responsiveness to house dust mite challenge. Our study shows that reactivity to house dust mite was associated with high basal levels of TNF-expressing cutaneous Th17 T cells, and documents the presence of hub structures where Langerhans cells and T cells co-localised. Mechanistically, we identify expression of metallothioneins and transcriptional programmes encoding antioxidant defences across all skin cell types, that appear to protect against allergen-induced inflammation. Furthermore, single nucleotide polymorphisms in the MTIX gene are associated with patients who did not react to house dust mite, opening up possibilities for therapeutic interventions modulating metallothionein expression in atopic dermatitis

    In silico modelling of toll-like receptor signalling pathways in human epidermal keratinocytes allows for prediction of immune responses to encountered antigens.

    No full text
    The cutaneous environment plays a pivotal role in the regulation of immune responses. A key aspect of this immune mediation are the keratinocytes which make up a large proportion of the epidermal layers. Their position in the epidermis means that they are often the first cell type to detect the invasion of pathogenic antigens, and are therefore responsible for initiating signalling pathways which dictate the severity of an immune response by releasing proinflammatory cytokines such as IL-8. In particular, TLR2 is known to detect the microbe S. aureus, which is commonly found in the cutaneous microbiome, particularly of those with atopic dermatitis and is known as a common cause for cutaneous infection and inflammation. Although experimental work has been carried out regarding the effects of S. aureus in relation to skin inflammatory responses, it is often difficult to investigate these responses in detail at the molecular level, particularly when a number of different experimental conditions are needed. Taking a systems biology approach can bridge the gap between global immune responses and in-depth molecular observations by using mathematics to describe the TLR2 signalling pathways in response to microbial ligands and utilising the model to predict how immune regulation will change under different conditions. A systematic review of TLR signalling pathways in human epidermal keratinocytes allowed for a global view of the interactions mediating immune responses. From this, an ordinary differential equations model was constructed to allow for quantitative modelling and predictions of keratinocyte immune responses following exposure to S. aureus. Model parameterisation was conducted using a genetic algorithm with rank selection which had been thoroughly tested on multiple systems biology models. After parameterising the TLR2 model with detailed time course data, it was possible to predict changes in IL-8 immune response following a change to ligand dose. Similarly, the TLR2 model provided accurate predictions of keratinocyte IL-8 secretion following exposure to lysed S. aureus. By modifying the TLR2 model to include a feedback loop, it was also possible to replicate the oscillatory dynamics observed in data from keratinocytes in a Th2 cytokine environment, suggesting that in an atopic dermatitis-like environment, molecular feedback pathways may be sensitised to show an amplification in oscillatory dynamics

    Dataset in support of the Southampton doctoral thesis 'In silico modelling of toll-like receptor signalling pathways in human epidermal keratinocytes allows for prediction of immune responses to encountered antigens'

    No full text
    Dataset title : Keratinocyte IL-8 secretion following challenge with TLR2 ligands ELISA results from experiment exposing human epidermal keratinocytes to LTA, staphylococcus aureus, or staphylococcus epidermidis and Th2 cytokines, with IL-8 secretions measured over an extended time course.</span

    Table_4_Transcriptional programming of immunoregulatory responses in human Langerhans cells.csv

    No full text
    Human epidermal Langerhans cells (LCs) maintain immune homeostasis in the skin. To examine transcriptional programming of human primary LCs during homeostasis, we performed scRNA-seq analysis of LCs before and after migration from the epidermis, coupled with functional assessment of their regulatory T cell priming capabilities. The analysis revealed that steady-state LCs exist in a continuum of maturation states and upregulate antigen presentation genes along with an immunoregulatory module including the genes IDO1, LGALS1, LAMTOR1, IL4I, upon their migration. The migration-induced transition in genomic state is accompanied by the ability of LCs to more efficiently prime regulatory T cell responses in co-culture assays. Computational analyses of the scRNAseq datasets using SCENIC and Partial Information Decomposition in Context identified a set of migration-induced transcription factors including IRF4, KLF6 and RelB as key nodes within a immunoregulatory gene regulatory network. These findings support a model in which efficient priming of immunoregulatory responses by LCs is dependent on coordinated upregulation of a migration-coupled maturation program with a immunoregulation-promoting genomic module.</p

    Table_5_Transcriptional programming of immunoregulatory responses in human Langerhans cells.csv

    No full text
    Human epidermal Langerhans cells (LCs) maintain immune homeostasis in the skin. To examine transcriptional programming of human primary LCs during homeostasis, we performed scRNA-seq analysis of LCs before and after migration from the epidermis, coupled with functional assessment of their regulatory T cell priming capabilities. The analysis revealed that steady-state LCs exist in a continuum of maturation states and upregulate antigen presentation genes along with an immunoregulatory module including the genes IDO1, LGALS1, LAMTOR1, IL4I, upon their migration. The migration-induced transition in genomic state is accompanied by the ability of LCs to more efficiently prime regulatory T cell responses in co-culture assays. Computational analyses of the scRNAseq datasets using SCENIC and Partial Information Decomposition in Context identified a set of migration-induced transcription factors including IRF4, KLF6 and RelB as key nodes within a immunoregulatory gene regulatory network. These findings support a model in which efficient priming of immunoregulatory responses by LCs is dependent on coordinated upregulation of a migration-coupled maturation program with a immunoregulation-promoting genomic module.</p

    DataSheet_1_Transcriptional programming of immunoregulatory responses in human Langerhans cells.pdf

    No full text
    Human epidermal Langerhans cells (LCs) maintain immune homeostasis in the skin. To examine transcriptional programming of human primary LCs during homeostasis, we performed scRNA-seq analysis of LCs before and after migration from the epidermis, coupled with functional assessment of their regulatory T cell priming capabilities. The analysis revealed that steady-state LCs exist in a continuum of maturation states and upregulate antigen presentation genes along with an immunoregulatory module including the genes IDO1, LGALS1, LAMTOR1, IL4I, upon their migration. The migration-induced transition in genomic state is accompanied by the ability of LCs to more efficiently prime regulatory T cell responses in co-culture assays. Computational analyses of the scRNAseq datasets using SCENIC and Partial Information Decomposition in Context identified a set of migration-induced transcription factors including IRF4, KLF6 and RelB as key nodes within a immunoregulatory gene regulatory network. These findings support a model in which efficient priming of immunoregulatory responses by LCs is dependent on coordinated upregulation of a migration-coupled maturation program with a immunoregulation-promoting genomic module.</p

    Table_1_Transcriptional programming of immunoregulatory responses in human Langerhans cells.xlsx

    No full text
    Human epidermal Langerhans cells (LCs) maintain immune homeostasis in the skin. To examine transcriptional programming of human primary LCs during homeostasis, we performed scRNA-seq analysis of LCs before and after migration from the epidermis, coupled with functional assessment of their regulatory T cell priming capabilities. The analysis revealed that steady-state LCs exist in a continuum of maturation states and upregulate antigen presentation genes along with an immunoregulatory module including the genes IDO1, LGALS1, LAMTOR1, IL4I, upon their migration. The migration-induced transition in genomic state is accompanied by the ability of LCs to more efficiently prime regulatory T cell responses in co-culture assays. Computational analyses of the scRNAseq datasets using SCENIC and Partial Information Decomposition in Context identified a set of migration-induced transcription factors including IRF4, KLF6 and RelB as key nodes within a immunoregulatory gene regulatory network. These findings support a model in which efficient priming of immunoregulatory responses by LCs is dependent on coordinated upregulation of a migration-coupled maturation program with a immunoregulation-promoting genomic module.</p

    Table_2_Transcriptional programming of immunoregulatory responses in human Langerhans cells.csv

    No full text
    Human epidermal Langerhans cells (LCs) maintain immune homeostasis in the skin. To examine transcriptional programming of human primary LCs during homeostasis, we performed scRNA-seq analysis of LCs before and after migration from the epidermis, coupled with functional assessment of their regulatory T cell priming capabilities. The analysis revealed that steady-state LCs exist in a continuum of maturation states and upregulate antigen presentation genes along with an immunoregulatory module including the genes IDO1, LGALS1, LAMTOR1, IL4I, upon their migration. The migration-induced transition in genomic state is accompanied by the ability of LCs to more efficiently prime regulatory T cell responses in co-culture assays. Computational analyses of the scRNAseq datasets using SCENIC and Partial Information Decomposition in Context identified a set of migration-induced transcription factors including IRF4, KLF6 and RelB as key nodes within a immunoregulatory gene regulatory network. These findings support a model in which efficient priming of immunoregulatory responses by LCs is dependent on coordinated upregulation of a migration-coupled maturation program with a immunoregulation-promoting genomic module.</p

    Table_6_Transcriptional programming of immunoregulatory responses in human Langerhans cells.csv

    No full text
    Human epidermal Langerhans cells (LCs) maintain immune homeostasis in the skin. To examine transcriptional programming of human primary LCs during homeostasis, we performed scRNA-seq analysis of LCs before and after migration from the epidermis, coupled with functional assessment of their regulatory T cell priming capabilities. The analysis revealed that steady-state LCs exist in a continuum of maturation states and upregulate antigen presentation genes along with an immunoregulatory module including the genes IDO1, LGALS1, LAMTOR1, IL4I, upon their migration. The migration-induced transition in genomic state is accompanied by the ability of LCs to more efficiently prime regulatory T cell responses in co-culture assays. Computational analyses of the scRNAseq datasets using SCENIC and Partial Information Decomposition in Context identified a set of migration-induced transcription factors including IRF4, KLF6 and RelB as key nodes within a immunoregulatory gene regulatory network. These findings support a model in which efficient priming of immunoregulatory responses by LCs is dependent on coordinated upregulation of a migration-coupled maturation program with a immunoregulation-promoting genomic module.</p

    Table_3_Transcriptional programming of immunoregulatory responses in human Langerhans cells.csv

    No full text
    Human epidermal Langerhans cells (LCs) maintain immune homeostasis in the skin. To examine transcriptional programming of human primary LCs during homeostasis, we performed scRNA-seq analysis of LCs before and after migration from the epidermis, coupled with functional assessment of their regulatory T cell priming capabilities. The analysis revealed that steady-state LCs exist in a continuum of maturation states and upregulate antigen presentation genes along with an immunoregulatory module including the genes IDO1, LGALS1, LAMTOR1, IL4I, upon their migration. The migration-induced transition in genomic state is accompanied by the ability of LCs to more efficiently prime regulatory T cell responses in co-culture assays. Computational analyses of the scRNAseq datasets using SCENIC and Partial Information Decomposition in Context identified a set of migration-induced transcription factors including IRF4, KLF6 and RelB as key nodes within a immunoregulatory gene regulatory network. These findings support a model in which efficient priming of immunoregulatory responses by LCs is dependent on coordinated upregulation of a migration-coupled maturation program with a immunoregulation-promoting genomic module.</p
    corecore