Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
J Invest Dermatol ; 143(7): 1257-1267.e10, 2023 07.
Article in English | MEDLINE | ID: mdl-36736996

ABSTRACT

Keratinocytes (KCs) form the outer epithelial barrier of the body, protecting against invading pathogens. Mice lacking the IL-17RA or both IL-17A and IL-17F develop spontaneous Staphylococcusaureus skin infections. We found a marked expansion of T17 cells, comprised of RORγt-expressing γδ T cells and T helper 17 cells in the skin-draining lymph nodes of these mice. Contradictory to previous suggestions, this expansion was not a result of a direct negative feedback loop because we found no expansion of T17 cells in mice lacking IL-17 signaling specifically in T cells. Instead, we found that the T17 expansion depended on the microbiota and was observed only when KCs were deficient for IL-17RA signaling. Indeed, mice that lack IL-17RA only in KCs showed an increased susceptibility to experimental epicutaneous infection with S. aureus together with an accumulation of IL-17A-producing γδ T cells. We conclude that deficiency of IL-17RA on KCs leads to microbiota dysbiosis in the skin, which triggers the expansion of IL-17A-producing T cells. Our data show that KCs are the primary target cells of IL-17A and IL-17F, coordinating the defense against microbial invaders in the skin.


Subject(s)
Interleukin-17 , Staphylococcus aureus , Mice , Animals , Mice, Knockout , Skin , Keratinocytes , Mice, Inbred C57BL
2.
J Invest Dermatol ; 142(1): 166-178.e8, 2022 01.
Article in English | MEDLINE | ID: mdl-34237339

ABSTRACT

Sodium can accumulate in the skin at concentrations exceeding serum levels. A high sodium environment can lead to pathogenic T helper 17 cell expansion. Psoriasis is a chronic inflammatory skin disease in which IL-17‒producing T helper 17 cells play a crucial role. In an observational study, we measured skin sodium content in patients with psoriasis and in age-matched healthy controls by Sodium-23 magnetic resonance imaging. Patients with PASI > 5 showed significantly higher sodium and water content in the skin but not in other tissues than those with lower PASI or healthy controls. Skin sodium concentrations measured by Sodium-23 spectroscopy or by atomic absorption spectrometry in ashed-skin biopsies verified the findings with Sodium-23 magnetic resonance imaging. In vitro T helper 17 cell differentiation of naive CD4+ cells from patients with psoriasis markedly induced IL-17A expression under increased sodium chloride concentrations. The imiquimod-induced psoriasis mouse model replicated the human findings. Extracellular tracer Chromium-51-EDTA measurements in imiquimod- and sham-treated skin showed similar extracellular volumes, rendering excessive water of intracellular origin. Chronic genetic IL-17A‒driven psoriasis mouse models underlined the role of IL-17A in dermal sodium accumulation and inflammation. Our data describe skin sodium as a pathophysiological feature of psoriasis, which could open new avenues for its treatment.


Subject(s)
Interleukin-17/metabolism , Psoriasis/metabolism , Skin/metabolism , Sodium/analysis , Th17 Cells/immunology , Animals , Cell Differentiation , Cells, Cultured , Humans , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Severity of Illness Index , Skin/pathology , Sodium Chloride/metabolism , Spectrophotometry, Atomic , Spectrum Analysis
3.
Proc Natl Acad Sci U S A ; 118(38)2021 09 21.
Article in English | MEDLINE | ID: mdl-34526403

ABSTRACT

The spleen contains phenotypically and functionally distinct conventional dendritic cell (cDC) subpopulations, termed cDC1 and cDC2, which each can be divided into several smaller and less well-characterized subsets. Despite advances in understanding the complexity of cDC ontogeny by transcriptional programming, the significance of posttranslational modifications in controlling tissue-specific cDC subset immunobiology remains elusive. Here, we identified the cell-surface-expressed A-disintegrin-and-metalloproteinase 10 (ADAM10) as an essential regulator of cDC1 and cDC2 homeostasis in the splenic marginal zone (MZ). Mice with a CD11c-specific deletion of ADAM10 (ADAM10ΔCD11c) exhibited a complete loss of splenic ESAMhi cDC2A because ADAM10 regulated the commitment, differentiation, and survival of these cells. The major pathways controlled by ADAM10 in ESAMhi cDC2A are Notch, signaling pathways involved in cell proliferation and survival (e.g., mTOR, PI3K/AKT, and EIF2 signaling), and EBI2-mediated localization within the MZ. In addition, we discovered that ADAM10 is a molecular switch regulating cDC2 subset heterogeneity in the spleen, as the disappearance of ESAMhi cDC2A in ADAM10ΔCD11c mice was compensated for by the emergence of a Clec12a+ cDC2B subset closely resembling cDC2 generally found in peripheral lymph nodes. Moreover, in ADAM10ΔCD11c mice, terminal differentiation of cDC1 was abrogated, resulting in severely reduced splenic Langerin+ cDC1 numbers. Next to the disturbed splenic cDC compartment, ADAM10 deficiency on CD11c+ cells led to an increase in marginal metallophilic macrophage (MMM) numbers. In conclusion, our data identify ADAM10 as a molecular hub on both cDC and MMM regulating their transcriptional programming, turnover, homeostasis, and ability to shape the anatomical niche of the MZ.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Dendritic Cells/metabolism , Membrane Proteins/metabolism , ADAM10 Protein/physiology , Amyloid Precursor Protein Secretases/physiology , Animals , Antigen-Presenting Cells/metabolism , CD11c Antigen/metabolism , Cell Differentiation , Cell Proliferation , Female , Homeostasis , Lymphoid Tissue/metabolism , Macrophages/metabolism , Male , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Signal Transduction , Spleen/cytology , Spleen/metabolism
4.
Article in English | MEDLINE | ID: mdl-33203651

ABSTRACT

OBJECTIVE: To assess the impact of laquinimod treatment on monocytes and to investigate the underlying immunomodulatory mechanisms in MS. METHODS: In this cross-sectional study, we performed in vivo and in vitro analyses of cluster of differentiation (CD14+) monocytes isolated from healthy donors (n = 15), untreated (n = 13), and laquinimod-treated patients with MS (n = 14). Their frequency and the expression of surface activation markers were assessed by flow cytometry and the viability by calcein staining. Cytokine concentrations in the supernatants of lipopolysaccharide (LPS)-stimulated monocytes were determined by flow cytometry. The messenger ribonucleic acid (mRNA) expression level of genes involved in cytokine expression was measured by quantitative PCR. The LPS-mediated nuclear factor kappa-light-chain-enhancer of activated B-cell (NF-κB) activation was determined by the quantification of the phosphorylation level of the p65 subunit. Laquinimod-treated monocytes were cocultured with CD4+ T cells, and the resulting cytokine production was analyzed by flow cytometry after intracellular cytokine staining. The interleukin (IL)-17A concentration of the supernatant was assessed by ELISA. RESULTS: Laquinimod did not alter the frequency or viability of circulating monocytes, but led to an upregulation of CD86 expression. LPS-stimulated monocytes of laquinimod-treated patients with MS secreted less IL-1ß following a downregulation of IL-1ß gene expression. Phosphorylation levels of the NF-κB p65 subunit were reduced after laquinimod treatment, indicating a laquinimod-associated inhibition of the NF-κB pathway. T cells primed with laquinimod-treated monocytes differentiated significantly less into IL-17A-producing T helper (Th)-17 cells. CONCLUSIONS: Our findings suggest that inhibited NF-κB signaling and downregulation of IL-1ß expression in monocytes contributes to the immunomodulatory effects of laquinimod and that the impairment of Th17 polarization might mediate its disease-modifying activity in MS.


Subject(s)
Interleukin-1beta/immunology , Monocytes/drug effects , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Quinolones/therapeutic use , Th17 Cells/immunology , Adult , Cross-Sectional Studies , Female , Humans , Interleukin-1beta/metabolism , Lymphocyte Activation/drug effects , Male , Middle Aged , Monocytes/immunology , Multiple Sclerosis, Relapsing-Remitting/immunology , Signal Transduction/drug effects
5.
Acta Neuropathol ; 140(4): 549-567, 2020 10.
Article in English | MEDLINE | ID: mdl-32651669

ABSTRACT

The proinflammatory cytokine interleukin 1 (IL-1) is crucially involved in the pathogenesis of multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Herein, we studied the role of IL-1 signaling in blood-brain barrier (BBB) endothelial cells (ECs), astrocytes and microglia for EAE development, using mice with the conditional deletion of its signaling receptor IL-1R1. We found that IL-1 signaling in microglia and astrocytes is redundant for the development of EAE, whereas the IL-1R1 deletion in BBB-ECs markedly ameliorated disease severity. IL-1 signaling in BBB-ECs upregulated the expression of the adhesion molecules Vcam-1, Icam-1 and the chemokine receptor Darc, all of which have been previously shown to promote CNS-specific inflammation. In contrast, IL-1R1 signaling suppressed the expression of the stress-responsive heme catabolizing enzyme heme oxygenase-1 (HO-1) in BBB-ECs, promoting disease progression via a mechanism associated with deregulated expression of the IL-1-responsive genes Vcam1, Icam1 and Ackr1 (Darc). Mechanistically, our data emphasize a functional crosstalk of BBB-EC IL-1 signaling and HO-1, controlling the transcription of downstream proinflammatory genes promoting the pathogenesis of autoimmune neuroinflammation.


Subject(s)
Blood-Brain Barrier/enzymology , Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelial Cells/enzymology , Heme Oxygenase-1/metabolism , Inflammation/immunology , Interleukin-1/immunology , Animals , Blood-Brain Barrier/immunology , Encephalomyelitis, Autoimmune, Experimental/enzymology , Gene Expression Regulation/immunology , Mice , Mice, Inbred C57BL , Signal Transduction/immunology
6.
Int J Mol Sci ; 21(3)2020 Jan 21.
Article in English | MEDLINE | ID: mdl-31973112

ABSTRACT

During the last decades, high-throughput assessment of gene expression in patient tissues using microarray technology or RNA-Seq took center stage in clinical research. Insights into the diversity and frequency of transcripts in healthy and diseased conditions provide valuable information on the cellular status in the respective tissues. Growing with the technique, the bioinformatic analysis toolkit reveals biologically relevant pathways which assist in understanding basic pathophysiological mechanisms. Conventional classification systems of inflammatory skin diseases rely on descriptive assessments by pathologists. In contrast to this, molecular profiling may uncover previously unknown disease classifying features. Thereby, treatments and prognostics of patients may be improved. Furthermore, disease models in basic research in comparison to the human disease can be directly validated. The aim of this article is not only to provide the reader with information on the opportunities of these techniques, but to outline potential pitfalls and technical limitations as well. Major published findings are briefly discussed to provide a broad overview on the current findings in transcriptomics in inflammatory skin diseases.


Subject(s)
Dermatitis, Atopic/metabolism , Psoriasis/metabolism , Skin Diseases/metabolism , Transcriptome , Computational Biology , Databases, Factual , Dermatitis, Atopic/genetics , Gene Expression Profiling/methods , Humans , Prognosis , Psoriasis/genetics , RNA-Seq/methods , Skin/metabolism , Skin Diseases/classification , Skin Diseases/genetics
7.
Nat Commun ; 10(1): 5722, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31844089

ABSTRACT

IL-17-producing CD8+ (Tc17) cells are enriched in active lesions of patients with multiple sclerosis (MS), suggesting a role in the pathogenesis of autoimmunity. Here we show that amelioration of MS by dimethyl fumarate (DMF), a mechanistically elusive drug, associates with suppression of Tc17 cells. DMF treatment results in reduced frequency of Tc17, contrary to Th17 cells, and in a decreased ratio of the regulators RORC-to-TBX21, along with a shift towards cytotoxic T lymphocyte gene expression signature in CD8+ T cells from MS patients. Mechanistically, DMF potentiates the PI3K-AKT-FOXO1-T-BET pathway, thereby limiting IL-17 and RORγt expression as well as STAT5-signaling in a glutathione-dependent manner. This results in chromatin remodeling at the Il17 locus. Consequently, T-BET-deficiency in mice or inhibition of PI3K-AKT, STAT5 or reactive oxygen species prevents DMF-mediated Tc17 suppression. Overall, our data disclose a DMF-AKT-T-BET driven immune modulation and suggest putative therapy targets in MS and beyond.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Dimethyl Fumarate/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Multiple Sclerosis/drug therapy , Adolescent , Adult , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dimethyl Fumarate/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Immunosuppressive Agents , Interleukin-17/immunology , Interleukin-17/metabolism , Longitudinal Studies , Male , Mice , Middle Aged , Multiple Sclerosis/blood , Multiple Sclerosis/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Treatment Outcome , Young Adult
8.
Nat Commun ; 10(1): 4877, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31653831

ABSTRACT

The interaction between the mammalian host and its resident gut microbiota is known to license adaptive immune responses. Nutritional constituents strongly influence composition and functional properties of the intestinal microbial communities. Here, we report that omission of a single essential amino acid - tryptophan - from the diet abrogates CNS autoimmunity in a mouse model of multiple sclerosis. Dietary tryptophan restriction results in impaired encephalitogenic T cell responses and is accompanied by a mild intestinal inflammatory response and a profound phenotypic shift of gut microbiota. Protective effects of dietary tryptophan restriction are abrogated in germ-free mice, but are independent of canonical host sensors of intracellular tryptophan metabolites. We conclude that dietary tryptophan restriction alters metabolic properties of gut microbiota, which in turn have an impact on encephalitogenic T cell responses. This link between gut microbiota, dietary tryptophan and adaptive immunity may help to develop therapeutic strategies for protection from autoimmune neuroinflammation.


Subject(s)
Autoimmunity/immunology , Diet , Encephalomyelitis, Autoimmune, Experimental/immunology , Gastrointestinal Microbiome/immunology , T-Lymphocytes/immunology , Tryptophan , Animals , Dietary Proteins , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/microbiology , Gastrointestinal Microbiome/genetics , Mice , Multiple Sclerosis , RNA, Ribosomal, 16S/genetics
9.
JCI Insight ; 4(22)2019 11 14.
Article in English | MEDLINE | ID: mdl-31622280

ABSTRACT

The transcriptional activator IκBζ is a key regulator of psoriasis, but which cells mediate its pathogenic effect remains unknown. Here we found that IκBζ expression in keratinocytes triggers not only skin lesions but also systemic inflammation in mouse psoriasis models. Specific depletion of IκBζ in keratinocytes was sufficient to suppress the induction of imiquimod- or IL-36-mediated psoriasis. Moreover, IκBζ ablation in keratinocytes prevented the onset of psoriatic lesions and systemic inflammation in keratinocyte-specific IL-17A-transgenic mice. Mechanistically, this psoriasis protection was mediated by IκBζ deficiency in keratinocytes abrogating the induction of specific proinflammatory target genes, including Cxcl5, Cxcl2, Csf2, and Csf3, in response to IL-17A or IL-36. These IκBζ-dependent genes trigger the generation and recruitment of neutrophils and monocytes that are needed for skin inflammation. Consequently, our data uncover a surprisingly pivotal role of keratinocytes and keratinocyte-derived IκBζ as key mediators of psoriasis and psoriasis-related systemic inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Inflammation/metabolism , Keratinocytes/metabolism , Psoriasis/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Female , Interleukin-17/genetics , Interleukin-17/metabolism , Male , Mice , Mice, Transgenic , Skin/cytology , Skin/pathology
10.
Free Radic Biol Med ; 141: 338-347, 2019 09.
Article in English | MEDLINE | ID: mdl-31279969

ABSTRACT

Dimethyl fumarate (DMF) is widely used to treat the human autoimmune diseases multiple sclerosis (MS) and psoriasis. DMF causes short-term oxidative stress and activates the antioxidant response via the transcription factor Nrf2 but its immunosuppressive effect is not well understood. Immune cell activation depends on calcium signaling which itself is influenced by the cellular redox state. We therefore measured calcium, reactive oxygen species levels and glutathione content in lymphocytes from immunized mice before onset of experimental autoimmune encephalomyelitis, in peripheral blood mononuclear cells from MS patients treated with DMF, and in mouse splenocytes treated ex vivo with DMF. This demonstrated altered redox states and increased lymphocytic calcium levels in all model systems. DMF caused an immediate influx of calcium from the extracellular space, long-term increased cytosolic calcium levels and reduced calcium stored in intracellular stores. The DMF-elicited current had the electrophysiological characteristics of a transient receptor potential channel and the intracellular calcium levels were normalized by antagonists of TRPA1. Interestingly, the sarco/endoplasmic reticulum Ca2+-ATPase SERCA2b was downregulated but more active due to glutathionylation of the redox-sensitive cysteine 674. DMF therefore causes pleiotropic changes in cellular calcium homeostasis which are likely caused by redox-sensitive post-translational modifications. These changes probably contribute to its immunosuppressive effects.


Subject(s)
Dimethyl Fumarate/pharmacology , Multiple Sclerosis/drug therapy , Psoriasis/drug therapy , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , TRPA1 Cation Channel/genetics , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Glutathione/metabolism , Humans , Lymphocytes/drug effects , Mice , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Oxidation-Reduction/drug effects , Psoriasis/genetics , Psoriasis/pathology , Reactive Oxygen Species/metabolism
11.
Nat Commun ; 10(1): 2730, 2019 06 21.
Article in English | MEDLINE | ID: mdl-31227691

ABSTRACT

Recently our groups discovered lugdunin, a new cyclic peptide antibiotic that inhibits Staphylococcus aureus epithelial colonization in humans and rodents. In this work, we analyzed its immuno-modulatory and antimicrobial potential as a single agent or in combination with other microbiota- or host-derived factors. We show that pretreatment of primary human keratinocytes or mouse skin with lugdunin in combination with microbiota-derived factors results in a significant reduction of S. aureus colonization. Moreover, lugdunin increases expression and release of LL-37 and CXCL8/MIP-2 in human keratinocytes and mouse skin, and results in the recruitment of monocytes and neutrophils in vivo, both by a TLR/MyD88-dependent mechanism. Interestingly, S. aureus elimination by lugdunin is additionally achieved by synergistic antimicrobial activity with LL-37 and dermcidin-derived peptides. In summary, our results indicate that lugdunin provides multi-level protection against S. aureus and may thus become a promising treatment option for S. aureus skin infections in the future.


Subject(s)
Anti-Bacterial Agents/pharmacology , Immunity, Innate/drug effects , Peptides, Cyclic/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Thiazolidines/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Antimicrobial Cationic Peptides/immunology , Cells, Cultured , Disease Models, Animal , Female , Humans , Keratinocytes/drug effects , Keratinocytes/immunology , Keratinocytes/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/drug effects , Microbiota/immunology , Peptides/immunology , Peptides, Cyclic/therapeutic use , Primary Cell Culture , Skin/drug effects , Skin/immunology , Skin/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Thiazolidines/therapeutic use , Cathelicidins
12.
J Invest Dermatol ; 139(5): 1110-1117, 2019 05.
Article in English | MEDLINE | ID: mdl-30684554

ABSTRACT

The pathology of psoriasis strongly depends on IL-17A. Monoclonal antibodies blocking either the cytokine or its receptor are among the most efficient treatments for psoriatic patients. Keratinocytes can be activated upon exposure to IL-17A and tumor necrosis factor-α and secrete secondary cytokines and chemokines in the inflamed skin. In psoriasis and its imiquimod-induced mouse model, a strong skin infiltration of neutrophils and inflammatory monocytes can be observed. However, to date, it is not clear how exactly those cellular populations are attracted to the skin and how they contribute to the pathogenesis of the disease. To define the crucial cell type responding to IL-17 and initiating the downstream pathology in psoriasis-like dermatitis, we used mice specifically lacking the IL-17 receptor (IL-17RA) in different cell types. Deletion of IL-17RA in T cells or myeloid had no impact on disease development. Only deletion of this receptor in keratinocytes reflected the full-body deletion of IL-17RA, resulting in strongly reduced dermatitis development. Imiquimod treatment of those IL-17 signaling-deficient mice maintained high monocytic infiltration but failed to attract neutrophils into the skin. We conclude that keratinocytes are a critical cellular target for IL-17A-mediated neutrophil attraction and psoriasis development.


Subject(s)
Adjuvants, Immunologic/pharmacology , Imiquimod/pharmacology , Interleukin-17/genetics , Psoriasis/genetics , Signal Transduction/drug effects , Animals , Biopsy, Needle , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , Flow Cytometry , Immunohistochemistry , Keratinocytes/drug effects , Keratinocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Psoriasis/chemically induced , Psoriasis/pathology , Random Allocation , Real-Time Polymerase Chain Reaction/methods , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/metabolism
13.
Gastroenterology ; 156(3): 692-707.e7, 2019 02.
Article in English | MEDLINE | ID: mdl-30315770

ABSTRACT

BACKGROUND & AIMS: The CYLD lysine 63 deubiquitinase gene (CYLD) encodes tumor suppressor protein that is mutated in familial cylindromatosus, and variants have been associated with Crohn disease (CD). Splice forms of CYLD that lack exons 7 and 8 regulate transcription factors and functions of immune cells. We examined the expression of splice forms of CYLD in colon tissues from patients with CD and their effects in mice. METHODS: We performed immunohistochemical analyses of colon tissues from patients with untreated CD and patients without inflammatory bowel diseases (controls). We obtained mice that expressed splice forms of CYLD (sCYLD mice) without or with SMAD7 (sCYLD/SMAD7 mice) from transgenes and CYLD-knockout mice (with or without transgenic expression of SMAD7) and performed endoscopic analyses. Colitis was induced in Rag1-/- mice by transfer of CD4+ CD62L+ T cells from C57/Bl6 or transgenic mice. T cells were isolated from mice and analyzed by flow cytometry and quantitative real-time polymerase chain reaction and intestinal tissues were analyzed by histology and immunohistochemistry. CYLD forms were expressed in mouse embryonic fibroblasts, primary T cells, and HEK293T cells, which were analyzed by immunoblot, mobility shift, and immunoprecipitation assays. RESULTS: The colonic lamina propria from patients with CD was infiltrated by T cells and had higher levels of sCYLD (but not full-length CYLD) and SMAD7 than tissues from controls. Incubation of mouse embryonic fibroblasts and T cells with transforming growth factor ß increased their production of sCYLD and decreased full-length CYLD. Transgenic expression of sCYLD and SMAD7 in T cells prevented the differentiation of regulatory T cells and T-helper type 17 cells and increased the differentiation of T-helper type 1 cells. The same effects were observed in colon tissues from sCYLD/SMAD7 mice but not in those from CYLD-knockout SMAD7 mice. The sCYLD mice had significant increases in the numbers of T-helper type 1 cells and CD44high CD62Llow memory-effector CD4+ T cells in the spleen and mesenteric lymph nodes compared with wild-type mice; sCYLD/SMAD7 mice had even larger increases. The sCYLD/SMAD7 mice spontaneously developed severe colitis, with infiltration of the colon by dendritic cells, neutrophils, macrophages, and CD4+ T cells and increased levels of Ifng, Il6, Il12a, Il23a, and Tnf mRNAs. Co-transfer of regulatory T cells from wild-type, but not from sCYLD/SMAD7, mice prevented the induction of colitis in Rag1-/- mice by CD4+ T cells. We found increased levels of poly-ubiquitinated SMAD7 in sCYLD CD4+ T cells. CYLD formed a nuclear complex with SMAD3, whereas sCYLD recruited SMAD7 to the nucleus, which inhibited the expression of genes regulated by SMAD3 and SMAD4. We found that sCYLD mediated lysine 63-linked ubiquitination of SMAD7. The sCYLD-SMAD7 complex inhibited transforming growth factor ß signaling in CD4+ T cells. CONCLUSIONS: Levels of the spliced form of CYLD are increased in colon tissues from patients with CD. sCYLD mediates ubiquitination and nuclear translocation of SMAD7 and thereby decreases transforming growth factor ß signaling in T cells. This prevents immune regulatory mechanisms and leads to colitis in mice.


Subject(s)
Crohn Disease/genetics , Crohn Disease/pathology , Cysteine Endopeptidases/genetics , Smad7 Protein/genetics , Ubiquitination/genetics , Animals , Biopsy, Needle , Deubiquitinating Enzyme CYLD , Disease Models, Animal , Flow Cytometry , Humans , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Random Allocation , Reference Values , Signal Transduction , Transforming Growth Factor beta1/genetics
14.
J Autoimmun ; 94: 110-121, 2018 11.
Article in English | MEDLINE | ID: mdl-30061013

ABSTRACT

NF-κB inducing kinase (NIK) is the key protein of the non-canonical NF-κB pathway and is important for the development of lymph nodes and other secondary immune organs. We elucidated the specific role of NIK in T cells using T-cell specific NIK-deficient (NIKΔT) mice. Despite showing normal development of lymphoid organs, NIKΔT mice were resistant to induction of CNS autoimmunity. T cells from NIKΔT mice were deficient in late priming, failed to up-regulate T-bet and to transmigrate into the CNS. Proteomic analysis of activated NIK-/- T cells showed de-regulated expression of proteins involved in the formation of the immunological synapse: in particular, proteins involved in cytoskeleton dynamics. In line with this we found that NIK-deficient T cells were hampered in phosphorylation of Zap70, LAT, AKT, ERK1/2 and PLCγ upon TCR engagement. Hence, our data disclose a hitherto unknown function of NIK in T-cell priming and differentiation.


Subject(s)
Actins/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Lymphocyte Activation , Protein Serine-Threonine Kinases/immunology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Actins/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Central Nervous System/immunology , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Profiling , Gene Expression Regulation , Lymph Nodes/immunology , Lymph Nodes/pathology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/immunology , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Peptide Fragments/administration & dosage , Phospholipase C gamma/genetics , Phospholipase C gamma/immunology , Phosphoproteins/genetics , Phosphoproteins/immunology , Primary Cell Culture , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Receptors, Antigen, T-Cell/genetics , Signal Transduction , Spleen/immunology , Spleen/pathology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , T-Lymphocytes/pathology , ZAP-70 Protein-Tyrosine Kinase/genetics , ZAP-70 Protein-Tyrosine Kinase/immunology , NF-kappaB-Inducing Kinase
15.
PLoS Biol ; 16(7): e2005380, 2018 07.
Article in English | MEDLINE | ID: mdl-29985916

ABSTRACT

Cofilin is an essential actin remodeling protein promoting depolymerization and severing of actin filaments. To address the relevance of cofilin for the development and function of T cells in vivo, we generated knock-in mice in which T-cell-specific nonfunctional (nf) cofilin was expressed instead of wild-type (WT) cofilin. Nf cofilin mice lacked peripheral αß T cells and showed a severe thymus atrophy. This was caused by an early developmental arrest of thymocytes at the double negative (DN) stage. Importantly, even though DN thymocytes expressed the TCRß chain intracellularly, they completely lacked TCRß surface expression. In contrast, nf cofilin mice possessed normal numbers of γδ T cells. Their functionality was confirmed in the γδ T-cell-driven, imiquimod (IMQ)-induced, psoriasis-like murine model. Overall, this study not only highlights the importance of cofilin for early αß T-cell development but also shows for the first time that an actin-binding protein is differentially involved in αß versus γδ T-cell development.


Subject(s)
Actin Depolymerizing Factors/metabolism , Actins/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Thymus Gland/metabolism , Actin Depolymerizing Factors/chemistry , Animals , Cell Movement , Gene Knock-In Techniques , Humans , Jurkat Cells , Mice , Mutation/genetics , Proline/metabolism , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , Thymocytes/metabolism
16.
J Mol Med (Berl) ; 96(8): 819-829, 2018 08.
Article in English | MEDLINE | ID: mdl-29959474

ABSTRACT

IL-17A and IL-17F share the highest sequence homology of the IL-17 family and signal via the same IL-17RA/RC receptor heterodimer. To better explore the expression of these two cytokines, we used a double reporter mouse strain (IL-17DR mice), where IL-17A expressing cells are marked by enhanced green fluorescent protein (eGFP) while red fluorescence protein (RFP) reports the expression of IL-17F. In steady state, we found that Th17 and γδ T cells only expressed IL-17A, while IL-17F expression was restricted to CD8 T cells (Tc17) and innate lymphoid cells (ILC type 3) of the gut. In experimental autoimmune encephalomyelitis, the vast majority of CNS-infiltrating Th17 cells expressed IL-17A but not IL-17F. In contrast, anti-CD3-induced, TGF-ß-driven Th17 cells in the gut expressed both of these IL-17 cytokines. In line with this, in vitro differentiation of Th17 cells in the presence of IL-1ß led primarily to IL-17A expressing T cells, while TGF-ß induced IL-17F co-expressing Th17 cells. Our results suggest that expression of IL-17F is associated with non-pathogenic T cells, pointing to a differential function of IL-17A versus IL-17F. KEY MESSAGES: Naïve mice: CD4+ T cells and γδ T cells express IL-17A, and Tc17 cells express IL-17F. Gut ILC3 show differential expression of IL17A and F. Th17 differentiation with TGF-ß1 induces IL-17A and F, whereas IL-1ß induced cells expressing IL-17A. Th17 cells in EAE in CNS express IL-17A only. Gut Th17 cells induced by anti-CD3 express IL-17A and F together as skin γδ T cells of IMQ-treated mice.


Subject(s)
Gene Expression , Interleukin-17/genetics , Th17 Cells/metabolism , Animals , Biomarkers , Cell Differentiation/immunology , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental , Immunophenotyping , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Transgenic , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/cytology , Th17 Cells/immunology
17.
Biochimie ; 153: 52-55, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29689289

ABSTRACT

Dihydroxycholesterols such as 7α,25-dihydroxysterols (7α,25-OHC) and 7α,27-OHC are generated from cholesterol by the enzymes CH25H, CYP7B1 and CYP27A1 in steady state but also in the context of inflammation. The G-protein coupled receptor (GPCR) Epstein-Barr virus-induced gene 2 (EBI2), also known as GPR183, senses these oxysterols and induces chemotactic migration of immune cells towards higher concentrations of these ligands. We recently showed that these ligands are upregulated in the CNS in experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis and that EBI2 enhanced early infiltration of encephalitogenic T cells into the CNS. In this short-review we discuss the role of dihydroxysterol-sensing by immune cells in neuroinflammation.


Subject(s)
Hydroxycholesterols/metabolism , Inflammation/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Chemotaxis , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Immunity, Cellular , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism
18.
Sci Rep ; 8(1): 5085, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572462

ABSTRACT

IL-22 is a potent pro-inflammatory cytokine upregulated in psoriasis and in other inflammatory diseases. The function of IL-22 is regulated by the soluble scavenging receptor, IL-22 binding protein (IL-22BP or IL-22RA2). However, the role and regulation of IL-22BP itself in the pathogenesis of inflammatory disease remain unclear. We used the TLR7 agonist Imiquimod (IMQ) to induce a psoriasis-like skin disease in mice and found a strong downregulation of IL-22BP in the affected skin as well as in the lymph nodes of animals treated with IMQ. We also analysed psoriatic skin of patients and compared this to skin of healthy donors. Interestingly, IL-22BP expression was similarly downregulated in skin biopsies of psoriasis patients compared to the skin of healthy donors. Since IL-22BP is expressed foremost in dendritic cells, we characterized its expression in monocyte-derived dendritic cells (MoDC) during maturation. In this way, we found Prostaglandin E2 (PGE2) to be a potent suppressor of IL-22BP expression in vitro. We conclude that regulation of IL-22BP by inflammatory mediators is an important step for the progression of inflammation in the skin and possibly also in other autoimmune diseases.


Subject(s)
Psoriasis/immunology , Receptors, Interleukin/immunology , Animals , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/pathology , Dinoprostone/analysis , Dinoprostone/immunology , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred BALB C , Psoriasis/pathology , Receptors, Interleukin/analysis , Skin/immunology , Skin/pathology
19.
JCI Insight ; 2(15)2017 Aug 03.
Article in English | MEDLINE | ID: mdl-28768912

ABSTRACT

GPCR expression was intensively studied in bulk cDNA of leukocyte populations, but limited data are available with respect to expression in individual cells. Here, we show a microfluidic-based single-cell GPCR expression analysis in primary T cells, myeloid cells, and endothelial cells under naive conditions and during experimental autoimmune encephalomyelitis, the mouse model of multiple sclerosis. We found that neuroinflammation induces characteristic changes in GPCR heterogeneity and patterning, and we identify various functionally relevant subgroups with specific GPCR profiles among spinal cord-infiltrating CD4 T cells, macrophages, microglia, or endothelial cells. Using GPCRs CXCR4, S1P1, and LPHN2 as examples, we show how this information can be used to develop new strategies for the functional modulation of Th17 cells and activated endothelial cells. Taken together, single-cell GPCR expression analysis identifies functionally relevant subpopulations with specific GPCR repertoires and provides a basis for the development of new therapeutic strategies in immune disorders.

20.
J Dermatol Sci ; 87(3): 221-227, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28633806

ABSTRACT

The cytokine IL-17 is now a target for an array of therapeutic monoclonal antibodies supposed to treat a variety of inflammatory diseases. The forerunner Secukinumab, an IL-17A neutralizing antibody, is meanwhile approved as first-line treatments for moderate-to-severe plaque psoriasis, and as second-line treatment for psoriatic arthritis and ankylosing spondylitis. Ixekizumab and Brodalumab, both also targeting the IL-17 pathway, were also recently approved by the FDA for plaque psoriasis. Using mice overexpressing IL-17A in a tissue of choice, we showed that the ectopic expression of this cytokine in keratinocytes resulted in a spontaneous and very strong form of psoriasis-like dermatitis. Interestingly, this model showed some typical comorbidities found in humans with psoriasis. In this review, we will discuss why IL-17 is a good target especially in psoriasis and what we learned from mouse models about its functions in pathological situations.


Subject(s)
Dermatitis/immunology , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Psoriasis/drug therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Dermatitis/pathology , Disease Models, Animal , Humans , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Transgenic , Molecular Targeted Therapy/methods , Psoriasis/immunology , Signal Transduction/immunology , Skin/immunology , Skin/pathology , Spondylitis, Ankylosing/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...