Interferon regulatory factor (IRF) family members impart cell-type specificity to toll-like receptor (TLR) signalling, and we recently identified a role for IRF6 in TLR2 signalling in epithelial cells. TLR3 has a well-characterized role in wound healing in the skin, and here, we examined TLR3-dependent IRF6 functions in human keratinocytes. Primary keratinocytes responded robustly to the TLR3 agonist poly(IC) with upregulation of mRNAs for interferon-β (IFN-β), the interleukin-12 (IL-12) family member IL-23p19 and the chemokines IL-8 and chemokine (C-C motif) ligand 5 (CCL5). Silencing of IRF6 expression enhanced poly(IC)-inducible IFN-β mRNA levels and inhibited poly(IC)-inducible IL-23p19 mRNA expression in primary keratinocytes. Consistent with these data, co-transfection of IRF6 increased poly(IC)-inducible IL-23p19 promoter activity, but inhibited poly(IC)-inducible IFN-β promoter activity in reporter assays. Surprisingly, poly(IC) did not regulate IL-12p40 expression in keratinocytes, suggesting that TLR3-inducible IL-23p19 may have an IL-23-independent function in these cells. The only other IL-12 family member that was strongly poly(IC) inducible was EBI3, which has not been shown to heterodimerize with IL-23p19. Both co-immunoprecipitation and proximity ligation assays revealed that IL-23p19 and EBI3 interact in cells. Co-expression of IL-23p19 and EBI3, as compared with IL-23p19 alone, resulted in increased levels of secreted IL-23p19, implying a functional role for this heterodimer. In summary, we report that IRF6 regulates a subset of TLR3 responses in human keratinocytes, including the production of a novel IL-12 family heterodimer (p19/EBI3). We propose that the TLR3-IRF6-p19/EBI3 axis may regulate keratinocyte and/or immune cell functions in the context of cell damage and wound healing in the skin.
The autoimmune disease type 1 diabetes is predominantly mediated by CD8+ cytotoxic T-cell destruction of islet beta cells, of which islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)206-214 is a dominant target antigen specificity. Previously, we found that a liposome-based antigen-specific immunotherapy encapsulating the CD4+ T-cell islet epitope 2.5mim together with the nuclear factor-κB inhibitor calcitriol induced regulatory T cells and protected from diabetes in NOD mice. Here we investigated whether the same system delivering IGRP206-214 could induce antigen-specific CD8+ T-cell-targeted immune regulation and delay diabetes. Subcutaneous administration of IGRP206-214 /calcitriol liposomes transiently activated and expanded IGRP-specific T-cell receptor transgenic 8.3 CD8+ T cells. Liposomal co-delivery of calcitriol was required to optimally suppress endogenous IGRP-specific CD8+ T-cell interferon-γ production and cytotoxicity. Concordantly, a short course of IGRP206-214 /calcitriol liposomes delayed diabetes progression and reduced insulitis. However, when IGRP206-214 /calcitriol liposomes were delivered together with 2.5mim /calcitriol liposomes, disease protection was not observed and the regulatory effect of 2.5mim /calcitriol liposomes was abrogated. Thus, tolerogenic liposomes that target either a dominant CD8+ or a CD4+ T-cell islet epitope can delay diabetes progression but combining multiple epitopes does not enhance protection.
Emerging evidence suggests that microbiome-host crosstalk regulates intestinal immune activity and predisposition to inflammatory bowel disease (IBD). NF-κB is a master regulator of immune function and a validated target for the treatment of IBD. Here, we identify five Clostridium strains that suppress immune-mediated NF-κB activation in epithelial cell lines, PBMCs, and gut epithelial organoids from healthy human subjects and patients with IBD. Cell-free culture supernatant from Clostridium bolteae AHG0001 strain, but not the reference C. bolteae BAA-613 strain, suppresses inflammatory responses and endoplasmic reticulum stress in gut epithelial organoids derived from Winnie mice. The in vivo responses to Clostridium bolteae AHG0001 and BAA-613 mirror the in vitro activity. Thus, using our in vitro screening of bacteria capable of suppressing NF-κB in the context of IBD and using an ex vivo organoid-based approach, we identify a strain capable of alleviating colitis in a relevant pre-clinical animal model of IBD.
Atopic dermatitis is a common pruritic and inflammatory skin disorder with unknown etiology. Most commonly occurring during early childhood, atopic dermatitis is associated with eczematous lesions and lichenification, in which the epidermis becomes hypertrophied resulting in thickening of the skin. In this study, we report an atopic dermatitis‐like pathophysiology results in a murine model following the expression of the high‐risk human papillomavirus (HPV) 16 oncoprotein E7 in keratinocytes under the keratin 14 promoter. We show that HPV16 E7 expression in the skin is associated with skin thickening, acanthosis and light spongiosis. Locally, HPV16 E7‐expressing skin secreted high levels of thymic stromal lymphopoietin (TSLP) and contained increased numbers of innate lymphoid cells (ILCs). High levels of circulating immunoglobulin E were associated with increased susceptibility to skin allergy in a model of cutaneous challenge, and to airway bronchiolar inflammation, enhanced airway goblet cell metaplasia and mucus production in a model of atopic march. Surprisingly, skin pathology occurred independently of T cells and mast cells. Thus, our findings suggest that the expression of a single HPV oncogene in the skin can drive the onset of atopic dermatitis‐like pathology through the induction of TSLP and type 2 ILC infiltration.
Abstract Objective Disturbances in immune regulation, intestinal microbial dysbiosis and intestinal inflammation characterize ankylosing spondylitis (AS), which is associated with RUNX3 loss-of-function variants. ZAP70 W163C mutant (SKG) mice have reduced ZAP70 signaling, spondyloarthritis and ileitis. At intestinal epithelial interfaces, lamina propria Foxp3 + regulatory T cells (Treg) and intraepithelial CD4 + CD8αα + TCRαβ + lymphocytes (CD4-IEL) control inflammation. TGF-β and retinoic acid (RA)-producing dendritic cells are required for induction of Treg and for CD4-IEL differentiation from CD4+ conventional or Treg precursors, with upregulation of Runx3 and suppression of ThPOK. We investigated Treg, CD4-IEL, ZAP70 and Runx3 in SKG mice and AS patients. Methods We compared ileal Treg and CD4-IEL numbers and differentiation in BALB/c and SKG mice, and with ZAP70 inhibition, and related differentially-expressed genes in terminal ileum to ChIP-seq-identified Runx3-regulated genes. We compared proportions of CD4-IEL in ileum and CD4 + 8 + T cells in blood of AS patients and healthy controls. Results ZAP70 W163C or ZAP70 inhibition prevented intestinal CD4-IEL but not Foxp3 + Treg differentiation in context of TGF-β and RA in vitro and in vivo, resulting in Runx3 and ThPOK dysregulation. CD4-IEL frequency and expression of tissue resident memory T-cell and Runx3-regulated genes was reduced in SKG intestine. Multiple under-expressed genes were shared with risk SNPs identified in human spondyloarthropathies. CD4-IEL were decreased in AS intestine. Double-positive T cells were reduced and Treg increased in AS peripheral blood. Conclusion High-affinity TCR-ZAP70 signalling is required for Runx3-mediated intestinal CD4-IEL differentiation from Treg. Genetically-encoded relative immunodeficiency of T cells underpins poor intestinal barrier control in mouse and human spondyloarthropathy. What is already known about this subject? Ankylosing spondylitis (AS) is associated with RUNX3 loss-of-function variants. Capacity of the AS T cell receptor repertoire to expand in response to infectious antigens is reduced. Foxp3 + regulatory T cells (Treg) are increased in AS intestine. ZAP70W163C mutant (SKG) mice have reduced ZAP70 signaling, spondyloarthritis (SpA) and ileitis. Intestinal epithelial Foxp3 + Treg and CD4 + CD8 + cytotoxic lymphocytes (CD4-IEL) control local inflammation. CD4-IEL differentiate from Treg, with upregulation of Runx3 and suppression of ThPOK transcription factors. What does this study add? High-affinity TCR-ZAP70 signalling is required for Runx3-mediated intestinal CD4-IEL differentiation from Treg Intestinal CD4-IEL and circulating CD4 + CD8 + T cells are reduced in AS while circulating Treg are increased. Impaired CD8 expression may be correctible by TNF inhibition in AS CD4 + T cells. Deficiencies of Runx3 and tissue-resident CD4-IEL link intestinal dysbiosis and inflammation in mouse and human SpA. How might this influence clinical practice or future developments? Genetically-encoded relative T immunodeficiency underpins poor intestinal barrier control in SpA
Abstract Natural regulatory T cells (Tregs) are present in high frequencies among tumor-infiltrating lymphocytes and in draining lymph nodes, supposedly facilitating tumor development. To investigate their role in controlling local immune responses, we analyzed intratumoral T cell accumulation and function in the presence or absence of Tregs. Tumors that grew in normal BALB/c mice injected with the 4T1 tumor cell line were highly infiltrated by Tregs, CD4 and CD8 cells, all having unique characteristics. Most infiltrating Tregs expressed low levels of CD25Rs and Foxp3. They did not proliferate even in the presence of IL-2 but maintained a strong suppressor activity. CD4 T cells were profoundly anergic and CD8 T cell proliferation and cytotoxicity were severely impaired. Depletion of Tregs modified the characteristics of tumor infiltrates. Tumors were initially invaded by activated CD4+CD25− T cells, which produced IL-2 and IFN-γ. This was followed by the recruitment of highly cytotoxic CD8+ T cells at tumor sites leading to tumor rejection. The beneficial effect of Treg depletion in tumor regression was abrogated when CD4 helper cells were also depleted. These findings indicate that the massive infiltration of tumors by Tregs prevents the development of a successful helper response. The Tregs in our model prevent Th cell activation and subsequent development of efficient CD8 T cell activity required for the control of tumor growth.
En presence de tumeur, des lymphocytes T regulateurs (Tregs) et effecteurs (Teffs) sont recrutes dans le ganglion drainant et la tumeur. Les Teffs ne menent au rejet tumoral qu’en l’absence de Tregs : les Tregs infiltrant la tumeur inhibent les Teffs CD8+ cytotoxiques via l’inhibition des CD4+ auxiliaires. Le recrutement des Tregs au niveau du ganglion drainant est un evenement precoce, a l’emergence de la tumeur : les Tregs, et surtout les Tregs memoires, ont une cinetique de reponse memoire contre les antigenes partages par les cellules normales et tumorales, alors que les Teffs ont une cinetique de reponse primaire contre les neoantigenes tumoraux. Si les Teffs ont un phenotype memoire contre la tumeur, ils ne sont alors plus sensibles a la suppression par les Tregs. En l’absence de Tregs, une bonne reponse effectrice necessite la presence d’un bon antigene tumoral (rejet tumoral). Mais un antigene trop immunodominant peut restreindre la reponse effectrice et mener a l’echappement des tumeurs (nouveau variant)
Abs to the prion protein (PrP) can protect against experimental prion infections, but efficient Ab responses are difficult to generate because PrP is expressed on many tissues and induces a strong tolerance. We previously showed that immunization of wild-type mice with PrP peptides and CpG oligodeoxynucleic acid overcomes tolerance and induces cellular and humoral responses to PrP. In this study, we compared Ab and T cell repertoires directed to PrP in wild-type and PrP knockout (Prnp o/o) C57BL/6 mice. Animals were immunized with mouse PrP-plasmid DNA or with 30-mer overlapping peptides either emulsified in CFA or CpG/IFA. In Prnp o/o mice, Abs raised by PrP-plasmid DNA immunization recognized only N-terminal PrP peptides; analyses of Ab responses after PrP peptide/CFA immunization allowed us to identify six distinct epitopes, five of which were also recognized by Abs raised by PrP peptides/CpG. By contrast, in wild-type mice, no Ab response was detected after PrP-plasmid DNA or peptide/CFA immunization. However, when using CpG, four C-terminal peptides induced Abs specific for distinct epitopes. Importantly, immune sera from Prnp o/o but not from wild-type mice bound cell surface PrP. Abs of IgG1 and IgG2b subclasses predominated in Prnp o/o mice while the strongest signals were for IgG2b in wild-type mice. Most anti-PrP Th cells were directed to a single epitope in both Prnp o/o and wild-type mice. We conclude that endogenous PrPC expression profoundly affects the Ab repertoire as B cells reactive for epitopes exposed on native PrPC are strongly tolerized. Implications for immunotherapy against prion diseases are discussed.