Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Rheumatology (Oxford) ; 61(6): 2682-2693, 2022 05 30.
Article in English | MEDLINE | ID: mdl-34559222

ABSTRACT

OBJECTIVE: SSc is a complex disease characterized by vascular abnormalities and inflammation culminating in hypoxia and excessive fibrosis. Previously, we identified chemokine (C-X-C motif) ligand 4 (CXCL4) as a novel predictive biomarker in SSc. Although CXCL4 is well-studied, the mechanisms driving its production are unclear. The aim of this study was to elucidate the mechanisms leading to CXCL4 production. METHODS: Plasmacytoid dendritic cells (pDCs) from 97 healthy controls and 70 SSc patients were cultured in the presence of hypoxia or atmospheric oxygen level and/or stimulated with several toll-like receptor (TLR) agonists. Further, pro-inflammatory cytokine production, CXCL4, hypoxia-inducible factor (HIF) -1α and HIF-2α gene and protein expression were assessed using ELISA, Luminex, qPCR, FACS and western blot assays. RESULTS: CXCL4 release was potentiated only when pDCs were simultaneously exposed to hypoxia and TLR9 agonist (P < 0.0001). Here, we demonstrated that CXCL4 production is dependent on the overproduction of mitochondrial reactive oxygen species (mtROS) (P = 0.0079) leading to stabilization of HIF-2α (P = 0.029). In addition, we show that hypoxia is fundamental for CXCL4 production by umbilical cord CD34 derived pDCs. CONCLUSION: TLR-mediated activation of immune cells in the presence of hypoxia underpins the pathogenic production of CXCL4 in SSc. Blocking either mtROS or HIF-2α pathways may therapeutically attenuate the contribution of CXCL4 to SSc and other inflammatory diseases driven by CXCL4.


Subject(s)
Platelet Factor 4/metabolism , Reactive Oxygen Species/metabolism , Scleroderma, Systemic , Toll-Like Receptor 9 , Basic Helix-Loop-Helix Transcription Factors/metabolism , Dendritic Cells/metabolism , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit
2.
Exp Dermatol ; 31(6): 962-969, 2022 06.
Article in English | MEDLINE | ID: mdl-35297512

ABSTRACT

Interleukin (IL)-12 and IL-23 are pro-inflammatory cytokines produced by dendritic cells (DCs) and associated with Psoriasis (Pso) and Psoriatic Arthritis (PsA) pathogenesis. Tofacitinib, a Janus kinase inhibitor, effectively suppresses inflammatory cascades downstream the IL-12/IL-23 axis in Pso and PsA patients. Here, we investigated whether Tofacitinib directly regulates IL-12/IL-23 production in DCs, and how this regulation reflects responses to Tofacitinib in Pso patients. We treated monocyte-derived dendritic cells and myeloid dendritic cells with Tofacitinib and stimulated cells with either lipopolysaccharide (LPS) or a combination of LPS and IFN-γ. We assessed gene expression by qPCR, obtained skin microarray and blood Olink data and clinical parameters of Pso patients treated with Tofacitinib from public data sets. Our results indicate that in DCs co-stimulated with LPS and IFN-γ, but not with LPS alone, Tofacitinib leads to the decreased expression of IL-23/IL-12 shared subunit IL12B (p40). In Tofacitinib-treated Pso patients, IL-12 expression and psoriasis area and severity index (PASI) are significantly reduced in patients with higher IFN-γ at baseline. These findings demonstrate for the first time that Tofacitinib suppresses IL-23/IL-12 shared subunit IL12B in DCs upon active IFN-γ signaling, and that Pso patients with higher IFN-γ baseline levels display improved clinical response after Tofacitinib treatment.


Subject(s)
Interferon-gamma , Interleukin-12 Subunit p40 , Janus Kinase Inhibitors , Piperidines , Psoriasis , Pyrimidines , Skin , Arthritis, Psoriatic/drug therapy , Dendritic Cells/immunology , Humans , Interferon-gamma/metabolism , Interleukin-12 Subunit p40/antagonists & inhibitors , Interleukin-12 Subunit p40/blood , Interleukin-12 Subunit p40/metabolism , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Lipopolysaccharides/immunology , Piperidines/pharmacology , Piperidines/therapeutic use , Psoriasis/drug therapy , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Skin/drug effects , Skin/immunology
3.
Immunity ; 38(2): 275-84, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23333074

ABSTRACT

Epidermal growth factor receptor (EGFR) is known to be critically involved in tissue development and homeostasis as well as in the pathogenesis of cancer. Here we showed that Foxp3(+) regulatory T (Treg) cells express EGFR under inflammatory conditions. Stimulation with the EGF-like growth factor Amphiregulin (AREG) markedly enhanced Treg cell function in vitro, and in a colitis and tumor vaccination model we showed that AREG was critical for efficient Treg cell function in vivo. In addition, mast cell-derived AREG fully restored optimal Treg cell function. These findings reveal EGFR as a component in the regulation of local immune responses and establish a link between mast cells and Treg cells. Targeting of this immune regulatory mechanism may contribute to the therapeutic successes of EGFR-targeting treatments in cancer patients.


Subject(s)
ErbB Receptors/immunology , Glycoproteins/immunology , Intercellular Signaling Peptides and Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Amphiregulin , Animals , Antibodies, Neutralizing/pharmacology , Cell Communication/immunology , Colitis/chemically induced , Colitis/immunology , Colitis/metabolism , Colitis/pathology , EGF Family of Proteins , ErbB Receptors/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gene Expression Regulation/drug effects , Glycoproteins/antagonists & inhibitors , Glycoproteins/genetics , Glycoproteins/pharmacology , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Lymphocyte Activation/drug effects , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Membrane Proteins/administration & dosage , Membrane Proteins/immunology , Mice , Mice, Transgenic , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
4.
Immunity ; 39(2): 259-71, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23973222

ABSTRACT

Stable Foxp3 expression is required for the development of functional regulatory T (Treg) cells. Here, we demonstrate that the expression of the transcription factor Foxp3 can be regulated through the polyubiquitination of multiple lysine residues, resulting in proteasome-mediated degradation. Expression of the deubiquitinase (DUB) USP7 was found to be upregulated and active in Treg cells, being associated with Foxp3 in the nucleus. Ectopic expression of USP7 decreased Foxp3 polyubiquitination and increased Foxp3 expression. Conversely, either treatment with DUB inhibitor or USP7 knockdown decreased endogenous Foxp3 protein expression and decreased Treg-cell-mediated suppression in vitro. Furthermore, in a murine adoptive-transfer-induced colitis model, either inhibition of DUB activity or USP7 knockdown in Treg cells abrogated their ability to resolve inflammation in vivo. Our data reveal a molecular mechanism in which rapid temporal control of Foxp3 expression in Treg cells can be regulated by USP7, thereby modulating Treg cell numbers and function.


Subject(s)
Colitis/immunology , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Ubiquitin Thiolesterase/metabolism , Adoptive Transfer , Animals , Cell Differentiation/immunology , Cell Line , DNA-Binding Proteins/genetics , Disease Models, Animal , Endopeptidases/metabolism , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , RNA Interference , RNA, Small Interfering , Ubiquitin Thiolesterase/genetics , Ubiquitin-Specific Peptidase 7 , Ubiquitination
5.
Eur J Immunol ; 50(1): 86-96, 2020 01.
Article in English | MEDLINE | ID: mdl-31713839

ABSTRACT

Non-Hodgkin orbital lymphoma (NHOL) and idiopathic orbital inflammation (IOI) are common orbital conditions with largely unknown pathophysiology that can be difficult to diagnose. In this study we aim to identify serum miRNAs associated with NHOL and IOI. We performed OpenArray® miRNA profiling in 33 patients and controls. Differentially expressed miRNAs were technically validated across technology platforms and replicated in an additional cohort of 32 patients and controls. We identified and independently validated a serum miRNA profile of NHOL that was remarkably similar to IOI and characterized by an increased expression of a cluster of eight miRNAs. Pathway enrichment analysis indicated that the miRNA-cluster is associated with immune-mediated pathways, which we supported by demonstrating the elevated expression of this cluster in serum of patients with other inflammatory conditions. The cluster contained miR-148a, a key driver of B-cell tolerance, and miR-365 that correlated with serum IgG and IgM concentrations. In addition, miR-29a and miR-223 were associated with blood lymphocyte and neutrophil populations, respectively. NHOL and IOI are characterized by an abnormal serum miRNA-cluster associated with immune pathway activation and linked to B cell and neutrophil dysfunction.


Subject(s)
Inflammation/immunology , Lymphoma, Non-Hodgkin/immunology , MicroRNAs/immunology , Orbital Diseases/immunology , Orbital Neoplasms/immunology , Adult , Aged , Female , Humans , Inflammation/genetics , Lymphoma, Non-Hodgkin/genetics , Male , Middle Aged , Orbital Diseases/genetics , Orbital Neoplasms/genetics
6.
Eur J Immunol ; 50(1): 119-129, 2020 01.
Article in English | MEDLINE | ID: mdl-31424086

ABSTRACT

Systemic sclerosis (SSc), systemic lupus erythematosus (SLE) and primary Sjögrens syndrome (pSS) are clinically distinct systemic autoimmune diseases (SADs) that share molecular pathways. We quantified the frequency of circulating immune-cells in 169 patients with these SADs and 44 healty controls (HC) using mass-cytometry and assessed the diagnostic value of these results. Alterations in the frequency of immune-cell subsets were present in all SADs compared to HC. Most alterations, including a decrease of CD56hi NK-cells in SSc and IgM+ Bcells in pSS, were disease specific; only a reduced frequency of plasmacytoid dendritic cells was common between all SADs Strikingly, hierarchical clustering of SSc patients identified 4 clusters associated with different clinical phenotypes, and 9 of the 12 cell subset-alterations in SSc were also present during the preclinical-phase of the disease. Additionally, we found a strong association between the use of prednisone and alterations in B-cell subsets. Although differences in immune-cell frequencies between these SADs are apparent, the discriminative value thereof is too low for diagnostic purposes. Within each disease, mass cytometry analyses revealed distinct patterns between endophenotypes. Given the lack of tools enabling early diagnosis of SSc, our results justify further research into the value of cellular phenotyping as a diagnostic aid.


Subject(s)
Flow Cytometry/methods , Lupus Erythematosus, Systemic/immunology , Scleroderma, Systemic/immunology , Sjogren's Syndrome/immunology , Adult , Aged , Female , Humans , Lupus Erythematosus, Systemic/diagnosis , Male , Middle Aged , Phenotype , Scleroderma, Systemic/diagnosis , Sjogren's Syndrome/diagnosis
7.
J Autoimmun ; 111: 102444, 2020 07.
Article in English | MEDLINE | ID: mdl-32284212

ABSTRACT

OBJECTIVE: To analyze how monocyte and macrophage exposure to CXCL4 induces inflammatory and fibrotic processes observed in Systemic sclerosis (SSc) patients. METHODS: In six independent experiments, monocytes of healthy controls (HC) and SSc patients were stimulated with CXCL4, TLR-ligands, IFNɑ or TGFß and the secretion of cytokines in the supernatant was assessed by multiplex immunoassays. PDGF-BB production by monocyte-derived macrophages was quantified using immunoassays. The number of monocytes and PDGF-BB in circulation was quantified in HC and SSc patients with the Sysmex XT-1800i haematology counter and immunoassays. Intracellular PDGF-BB was quantified in monocytes by Western blot. PDGF-receptor inhibition was achieved using siRNA-mediated knockdown or treatment with Crenolanib. The production of inflammatory mediators and extracellular matrix (ECM) components by dermal fibroblasts was analyzed by qPCR, ELISA and ECM deposition assays. RESULTS: SSc and HC monocytes released PDGF-BB upon stimulation with CXCL4. Conversely, TLR ligands, IFNɑ or TGFß did not induce PDGF-bb release. PDGF-BB plasma levels were significantly (P = 0.009) higher in diffuse SSc patients (n = 19), compared with HC (n = 21). In healthy dermal fibroblasts, PDGF-BB enhanced TNFɑ-induced expression of inflammatory cytokines and increased ECM production. Comparable results were observed in fibroblasts cultured in supernatant taken from macrophages stimulated with CXCL4. This effect was almost completely abrogated by inhibition of the PDGF-receptor using Crenolanib. CONCLUSION: Our findings demonstrate that CXCL4 can drive fibroblast activation indirectly via PDGF-BB production by myeloid cells. Hence, targeting PDGF-BB or CXCL4-induced PDGF-BB release could be clinically beneficial for patients with SSc.


Subject(s)
Becaplermin/metabolism , Fibroblasts/immunology , Inflammation/immunology , Macrophages/immunology , Monocytes/immunology , Platelet Factor 4/metabolism , Scleroderma, Systemic/immunology , Adult , Aged , Benzimidazoles/pharmacology , Cells, Cultured , Cytokines/metabolism , Female , Humans , Male , Middle Aged , Piperidines/pharmacology , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors
8.
Ann Rheum Dis ; 78(4): 529-538, 2019 04.
Article in English | MEDLINE | ID: mdl-30793699

ABSTRACT

BACKGROUND AND OBJECTIVE: Systemic sclerosis (SSc) is a severe autoimmune disease, in which the pathogenesis is dependent on both genetic and epigenetic factors. Altered gene expression in SSc monocytes, particularly of interferon (IFN)-responsive genes, suggests their involvement in SSc development. We investigated the correlation between epigenetic histone marks and gene expression in SSc monocytes. METHODS: Chromatin immunoprecipitation followed by sequencing (ChIPseq) for histone marks H3K4me3 and H3K27ac was performed on monocytes of nine healthy controls and 14 patients with SSc. RNA sequencing was performed in parallel to identify aberrantly expressed genes and their correlation with the levels of H3K4me3 and H3K27ac located nearby their transcription start sites. ChIP-qPCR assays were used to verify the role of bromodomain proteins, H3K27ac and STATs on IFN-responsive gene expression. RESULTS: 1046 and 534 genomic loci showed aberrant H3K4me3 and H3K27ac marks, respectively, in SSc monocytes. The expression of 381 genes was directly and significantly proportional to the levels of such chromatin marks present near their transcription start site. Genes correlated to altered histone marks were enriched for immune, IFN and antiviral pathways and presented with recurrent binding sites for IRF and STAT transcription factors at their promoters. IFNα induced the binding of STAT1 and STAT2 at the promoter of two of these genes, while blocking acetylation readers using the bromodomain BET family inhibitor JQ1 suppressed their expression. CONCLUSION: SSc monocytes have altered chromatin marks correlating with their IFN signature. Enzymes modulating these reversible marks may provide interesting therapeutic targets to restore monocyte homeostasis to treat or even prevent SSc.


Subject(s)
Epigenesis, Genetic , Histone Code/genetics , Monocytes/immunology , Scleroderma, Systemic/genetics , Adult , Aged , Azepines/pharmacology , Case-Control Studies , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/immunology , Female , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Histones/genetics , Humans , Interferon-alpha/immunology , Male , Middle Aged , Molecular Targeted Therapy/methods , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Scleroderma, Systemic/immunology , Triazoles/pharmacology
9.
J Autoimmun ; 89: 162-170, 2018 05.
Article in English | MEDLINE | ID: mdl-29371048

ABSTRACT

OBJECTIVE: MicroRNAs (miRNAs) are regulatory molecules, which have been addressed as potential biomarkers and therapeutic targets in rheumatic diseases. Here, we investigated the miRNA signature in the serum of systemic sclerosis (SSc) patients and we further assessed their expression in early stages of the disease. METHODS: The levels of 758 miRNAs were evaluated in the serum of 26 SSc patients as compared to 9 healthy controls by using an Openarray platform. Three miRNAs were examined in an additional cohort of 107 SSc patients and 24 healthy donors by single qPCR. MiR-483-5p expression was further analysed in the serum of patients with localized scleroderma (LoS) (n = 22), systemic lupus erythematosus (SLE) (n = 33) and primary Sjögren's syndrome (pSS) (n = 23). The function of miR-483-5p was examined by transfecting miR-483-5p into primary human dermal fibroblasts and pulmonary endothelial cells. RESULTS: 30 miRNAs were significantly increased in patients with SSc. Of these, miR-483-5p showed reproducibly higher levels in an independent SSc cohort and was also elevated in patients with preclinical-SSc symptoms (early SSc). Notably, miR-483-5p was not differentially expressed in patients with SLE or pSS, whereas it was up-regulated in LoS, indicating that this miRNA could be involved in the development of skin fibrosis. Consistently, miR-483-5p overexpression in fibroblasts and endothelial cells modulated the expression of fibrosis-related genes. CONCLUSIONS: Our findings showed that miR-483-5p is up-regulated in the serum of SSc patients, from the early stages of the disease onwards, and indicated its potential function as a fine regulator of fibrosis in SSc.


Subject(s)
Endothelial Cells/physiology , Fibroblasts/physiology , MicroRNAs/genetics , Scleroderma, Systemic/genetics , Skin/pathology , Adult , Aged , Cohort Studies , Female , Fibrosis , Genetic Testing , Humans , Male , Middle Aged , Up-Regulation
10.
Rheumatology (Oxford) ; 57(9): 1669-1674, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29873766

ABSTRACT

Objective: To investigate miRNA expression in relation to transcriptomic changes in plasmacytoid dendritic cells (pDCs) in SLE and APS. pDCs are major producers of IFNα in SLE and APS, and miRNAs are emerging as regulators of pDC activation. Methods: miRNA and mRNA expression were measured by OpenArray and RNA-sequencing in pDCs of SLE, SLE + APS (APS secondary to SLE) and primary APS (PAPS) patients. The miRNA profile of patients was compared with the miRNA pattern of TLR7-activated pDCs. Results: Among 131 miRNAs detected in pDCs, 35, 17 and 21 had a significantly lower level of expression in SLE, SLE + APS and PAPS patients, respectively, as compared with healthy controls (HC). Notably, the miRNA profile did not significantly differ between SLE and APS, but was driven by the presence or absence of an IFN signature. TLR7 stimulation induced a general downregulation of miRNAs, similar to the pattern observed in SLE and APS patients. miR-361-5p, miR-128-3p and miR-181a-2-3p expression was lower in patients with a high IFN signature (false discovery rate <0.05) as compared with patients with a low IFN signature and HCs. Pathway enrichment on the overlap of miRNA targets and upregulated genes from the RNAseq indicated that these miRNAs are involved in pDC activation and apoptosis. Conclusion: Lower miRNA expression in pDCs is shared between SLE, SLE + APS and PAPS and is related to the IFN signature. As pDCs are the alleged source of the IFN signature in these patients, a better understanding of the molecular mechanisms/pathways leading to pDC dysregulation in SLE and APS might open novel pathways for therapeutic intervention.


Subject(s)
Antiphospholipid Syndrome/genetics , Dendritic Cells/metabolism , Down-Regulation , Gene Expression Regulation , Lupus Erythematosus, Systemic/genetics , MicroRNAs/genetics , Toll-Like Receptor 7/genetics , Adult , Antiphospholipid Syndrome/metabolism , Dendritic Cells/pathology , Female , Humans , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Male , MicroRNAs/biosynthesis , RNA, Messenger/genetics , Toll-Like Receptor 7/biosynthesis
11.
J Immunol ; 187(5): 2302-9, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21804012

ABSTRACT

Exposure of cells to inflammatory cytokines induces the expression of three proteasome immunosubunits, two of which are encoded in the MHC class II region. The induced subunits replace their constitutive homologs in newly formed "so-called" immunoproteasomes. Immunosubunit incorporation enhances the proteasome's proteolytic activity and modifies the proteasome's cleavage-site preferences, which improves the generation of many MHC class I-presented peptides and shapes the fine specificity of pathogen-specific CD8 T cell responses. In this article, we report on a second effect of immunoproteasome formation on CD8 T cell responses. We show that mice deficient for the immunosubunits ß5i/low molecular mass polypeptide (LMP7) and ß2i/multicatalytic endopeptidase complex-like-1 develop early-stage multiorgan autoimmunity following irradiation and bone marrow transplantation. Disease symptoms are caused by CD8 T cells and are transferable into immunosubunit-deficient, RAG1-deficient mice. Moreover, using the human Type 1 Diabetes Genetics Consortium MHC dataset, we identified two single nucleotide polymorphisms within the ß5i/LMP7-encoding gene sequences, which were in strong linkage disequilibrium, as independent genetic risk factors for type 1 diabetes development in humans. Strikingly, these single nucleotide polymorphisms significantly enhanced the risk conferred by HLA haplotypes that were previously shown to predispose for type 1 diabetes. These data suggested that inflammation-induced immunosubunit expression in peripheral tissues constitutes a mechanism that prevents the development of CD8 T cell-mediated autoimmune diseases.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Genetic Predisposition to Disease , Humans , Linkage Disequilibrium , Mice , Mice, Knockout , Polymorphism, Single Nucleotide , Risk Factors
12.
RMD Open ; 9(3)2023 08.
Article in English | MEDLINE | ID: mdl-37532471

ABSTRACT

OBJECTIVES: To assess to what extent leflunomide (LEF) and hydroxychloroquine (HCQ) therapy in patients with primary Sjögren's syndrome (RepurpSS-I) targets type I IFN-associated responses and to study the potential of several interferon associated RNA-based and protein-based biomarkers to predict and monitor treatment. METHODS: In 21 patients treated with LEF/HCQ and 8 patients treated with placebo, blood was drawn at baseline, 8, 16 and 24 weeks. IFN-signatures based on RNA expression of five IFN-associated genes were quantified in circulating mononuclear cells and in whole blood. MxA protein levels were measured in whole blood, and protein levels of CXCL10 and Galectin-9 were quantified in serum. Differences between responders and non-responders were assessed and receiver operating characteristic analysis was used to determine the capacity of baseline expression and early changes (after 8 weeks of treatment) in biomarkers to predict treatment response at the clinical endpoint. RESULTS: IFN-signatures in peripheral blood mononuclear cell and whole blood decreased after 24 weeks of LEF/HCQ treatment, however, changes in IFN signatures only poorly correlated with changes in disease activity. In contrast to baseline IFN signatures, baseline protein concentrations of galectin-9 and decreases in circulating MxA and Galectin-9 were robustly associated with clinical response. Early changes in serum Galectin-9 best predicted clinical response at 24 weeks (area under the curve 0.90). CONCLUSIONS: LEF/HCQ combination therapy targets type-I IFN-associated proteins that are associated with strongly decreased B cell hyperactivity and disease activity. IFN-associated Galectin-9 is a promising biomarker for treatment prediction and monitoring in pSS patients treated with LEF/HCQ.


Subject(s)
Interferon Type I , Sjogren's Syndrome , Humans , Biomarkers , Hydroxychloroquine/therapeutic use , Interferon Type I/metabolism , Leflunomide/therapeutic use , Leukocytes, Mononuclear/metabolism , Proteins , RNA , Sjogren's Syndrome/diagnosis , Sjogren's Syndrome/drug therapy
13.
Arthritis Rheumatol ; 75(2): 279-292, 2023 02.
Article in English | MEDLINE | ID: mdl-36482877

ABSTRACT

OBJECTIVE: This study was undertaken to identify key disease pathways driving conventional dendritic cell (cDC) alterations in systemic sclerosis (SSc). METHODS: Transcriptomic profiling was performed on peripheral blood CD1c+ cDCs (cDC2s) isolated from 12 healthy donors and 48 patients with SSc, including all major disease subtypes. We performed differential expression analysis for the different SSc subtypes and healthy donors to uncover genes dysregulated in SSc. To identify biologically relevant pathways, we built a gene coexpression network using weighted gene correlation network analysis. We validated the role of key transcriptional regulators using chromatin immunoprecipitation (ChIP) sequencing and in vitro functional assays. RESULTS: We identified 17 modules of coexpressed genes in cDCs that correlated with SSc subtypes and key clinical traits, including autoantibodies, skin score, and occurrence of interstitial lung disease. A module of immunoregulatory genes was markedly down-regulated in patients with the diffuse SSc subtype characterized by severe fibrosis. Transcriptional regulatory network analysis performed on this module predicted nuclear receptor 4A (NR4A) subfamily genes (NR4A1, NR4A2, NR4A3) as the key transcriptional regulators of inflammation. Indeed, ChIP-sequencing analysis indicated that these NR4A members target numerous differentially expressed genes in SSc cDC2s. Inclusion of NR4A receptor agonists in culture-based experiments provided functional proof that dysregulation of NR4As affects cytokine production by cDC2s and modulates downstream T cell activation. CONCLUSION: NR4A1, NR4A2, and NR4A3 are important regulators of immunosuppressive and fibrosis-associated pathways in SSc cDCs. Thus, the NR4A family represents novel potential targets to restore cDC homeostasis in SSc.


Subject(s)
Nuclear Receptor Subfamily 4, Group A, Member 2 , Scleroderma, Systemic , Humans , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Gene Expression Regulation , Gene Expression , Scleroderma, Systemic/genetics , Fibrosis , Glycoproteins/metabolism , Antigens, CD1/genetics
14.
J Invest Dermatol ; 142(2): 402-413, 2022 02.
Article in English | MEDLINE | ID: mdl-34333017

ABSTRACT

Dermal fibroblasts are strategically positioned underneath the basal epidermis layer to support keratinocyte proliferation and extracellular matrix production. In inflammatory conditions, these fibroblasts produce cytokines and chemokines that promote the chemoattraction of immune cells into the dermis and the hyperplasia of the epidermis, two characteristic hallmarks of psoriasis. However, how dermal fibroblasts specifically contribute to psoriasis development remains largely uncharacterized. In this study, we investigated through which cytokines and signaling pathways dermal fibroblasts contribute to the inflammatory features of psoriatic skin. We show that dermal fibroblasts from lesional psoriatic skin are important producers of inflammatory mediators, including IL-6, CXCL8, and CXCL2. This increased cytokine production was found to be regulated by ZFP36 family members ZFP36, ZFP36L1, and ZFP36L2, RNA-binding proteins with mRNA-degrading properties. In addition, the expression of ZFP36 family proteins was found to be reduced in chronic inflammatory conditions that mimic psoriatic lesional skin. Collectively, these results indicate that dermal fibroblasts are important producers of cytokines in psoriatic skin and that reduced expression of ZFP36 members in psoriasis dermal fibroblasts contributes to their inflammatory phenotype.


Subject(s)
Butyrate Response Factor 1/metabolism , Fibroblasts/metabolism , Psoriasis/immunology , Transcription Factors/metabolism , Tristetraprolin/metabolism , Biopsy , Butyrate Response Factor 1/genetics , Case-Control Studies , Epidermis/immunology , Epidermis/metabolism , Epidermis/pathology , Gene Knockdown Techniques , Healthy Volunteers , Humans , Inflammation Mediators/metabolism , Keratinocytes/immunology , Keratinocytes/metabolism , Psoriasis/pathology , Transcription Factors/genetics , Tristetraprolin/genetics
15.
Cell Rep ; 38(1): 110189, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34986347

ABSTRACT

Fibrosis is a major cause of mortality worldwide, characterized by myofibroblast activation and excessive extracellular matrix deposition. Systemic sclerosis is a prototypic fibrotic disease in which CXCL4 is increased and strongly correlates with skin and lung fibrosis. Here we aim to elucidate the role of CXCL4 in fibrosis development. CXCL4 levels are increased in multiple inflammatory and fibrotic mouse models, and, using CXCL4-deficient mice, we demonstrate the essential role of CXCL4 in promoting fibrotic events in the skin, lungs, and heart. Overexpressing human CXCL4 in mice aggravates, whereas blocking CXCL4 reduces, bleomycin-induced fibrosis. Single-cell ligand-receptor analysis predicts CXCL4 to affect endothelial cells and fibroblasts. In vitro, we confirm that CXCL4 directly induces myofibroblast differentiation and collagen synthesis in different precursor cells, including endothelial cells, by stimulating endothelial-to-mesenchymal transition. Our findings identify a pivotal role of CXCL4 in fibrosis, further substantiating the potential role of neutralizing CXCL4 as a therapeutic strategy.


Subject(s)
Extracellular Matrix/pathology , Myofibroblasts/metabolism , Platelet Factor 4/metabolism , Pulmonary Fibrosis/pathology , Scleroderma, Systemic/pathology , Animals , Bleomycin/toxicity , Cell Line , Collagen/biosynthesis , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/physiology , Human Umbilical Vein Endothelial Cells , Humans , Lung/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/cytology , Pericytes/metabolism , Platelet Factor 4/genetics , Stromal Cells/cytology , Stromal Cells/metabolism
16.
Front Immunol ; 12: 701656, 2021.
Article in English | MEDLINE | ID: mdl-34413853

ABSTRACT

Primary Sjögren's syndrome (pSS) is a systemic autoimmune disease characterized by infiltration of the exocrine glands and prominent B cell hyperactivity. Considering the key role of monocytes in promoting B cell hyperactivity, we performed RNA-sequencing analysis of CD14+ monocytes from patients with pSS, non-Sjögren's sicca (nSS), and healthy controls (HC). We demonstrated that the transcriptomic profile of pSS patients is enriched in intermediate and non-classical monocyte profiles, and confirmed the increased frequency of non-classical monocytes in pSS patients by flow-cytometry analysis. Weighted gene co-expression network analysis identified four molecular signatures in monocytes from pSS patients, functionally annotated for processes related with translation, IFN-signaling, and toll-like receptor signaling. Systemic and local inflammatory features significantly correlated with the expression of these signatures. Furthermore, genes highly associated with clinical features in pSS were identified as hub-genes for each signature. Unsupervised hierarchical cluster analysis of the hub-genes identified four clusters of nSS and pSS patients, each with distinct inflammatory and transcriptomic profiles. One cluster showed a significantly higher percentage of pSS patients with higher prevalence of anti-SSA autoantibodies, interferon-score, and erythrocyte sedimentation rate compared to the other clusters. Finally, we showed that the identified transcriptomic differences in pSS monocytes were induced in monocytes of healthy controls by exposure to serum of pSS patients. Representative hub-genes of all four signatures were partially inhibited by interferon-α/ß receptor blockade, indicating that the circulating inflammatory mediators, including type I interferons have a significant contribution to the altered transcriptional profile of pSS-monocytes. Our study suggests that targeting key circulating inflammatory mediators, such as type I interferons, could offer new insights into the important pathways and mechanisms driving pSS, and holds promise for halting immunopathology in Sjögren's Syndrome.


Subject(s)
Inflammation/genetics , Monocytes/pathology , Sjogren's Syndrome/genetics , Sjogren's Syndrome/pathology , Transcriptome/genetics , Adult , Aged , Aged, 80 and over , Autoantibodies/genetics , Female , Humans , Inflammation Mediators/metabolism , Interferon Type I/genetics , Lipopolysaccharide Receptors/genetics , Male , Middle Aged , Signal Transduction/genetics , Toll-Like Receptors/genetics , Young Adult
17.
J Clin Med ; 10(3)2021 Jan 30.
Article in English | MEDLINE | ID: mdl-33573268

ABSTRACT

Compelling evidence shows the involvement of plasmacytoid dendritic cells (pDCs) in systemic sclerosis (SSc) pathogenesis. This study investigated whether microRNAs (miRNAs) are involved in the dysregulation of pDCs in SSc patients already at early stages. RNA from circulating pDCs was isolated from two independent cohorts of SSc patients with different disease phenotypes, and individuals with Raynaud's phenomenon, for microRNA profiling and RNA-sequencing analysis. Proteomic analysis was exploited to identify novel direct miRNA targets at the protein level. Twelve and fifteen miRNAs were differentially expressed in at least one group of patients compared to healthy controls in discovery cohort I and II, respectively. Of note, miR-126 and miR-139-5p were upregulated in both preclinical and definite SSc patients and correlated with the expression of type I interferon (IFN)-responsive genes. Toll-like receptor 9 (TLR9) stimulation of healthy pDCs upregulated the expression of both miRNAs, similarly to what was observed in patients. The proteomic analysis identified USP24 as a novel target of miR-139-5p. The expression level of USP24 was inversely correlated with miR-139-5p expression in SSc patients and induced by TLR9 stimulation in healthy pDCs. These findings demonstrated that the miRNA profile is altered in pDCs of SSc patients already at early stages of the disease and indicate their potential contribution to pDC activation observed in patients.

18.
Front Immunol ; 11: 2149, 2020.
Article in English | MEDLINE | ID: mdl-33042127

ABSTRACT

Fibrosis is a condition shared by numerous inflammatory diseases. Our incomplete understanding of the molecular mechanisms underlying fibrosis has severely hampered effective drug development. CXCL4 is associated with the onset and extent of fibrosis development in multiple inflammatory and fibrotic diseases. Here, we used monocyte-derived cells as a model system to study the effects of CXCL4 exposure on dendritic cell development by integrating 65 longitudinal and paired whole genome transcriptional and methylation profiles. Using data-driven gene regulatory network analyses, we demonstrate that CXCL4 dramatically alters the trajectory of monocyte differentiation, inducing a novel pro-inflammatory and pro-fibrotic phenotype mediated via key transcriptional regulators including CIITA. Importantly, these pro-inflammatory cells directly trigger a fibrotic cascade by producing extracellular matrix molecules and inducing myofibroblast differentiation. Inhibition of CIITA mimicked CXCL4 in inducing a pro-inflammatory and pro-fibrotic phenotype, validating the relevance of the gene regulatory network. Our study unveils that CXCL4 acts as a key secreted factor driving innate immune training and forming the long-sought link between inflammation and fibrosis.


Subject(s)
Dendritic Cells/cytology , Fibrosis/immunology , Gene Regulatory Networks , Inflammation/immunology , Platelet Factor 4/physiology , Transcriptome , Cells, Cultured , Cellular Reprogramming Techniques , DNA Methylation , Decision Trees , Decitabine/pharmacology , Fibroblasts , Fibrosis/genetics , Humans , Inflammation/genetics , Monocytes/cytology , Multidimensional Scaling Analysis , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/physiology , Poly I-C/pharmacology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , RNA-Seq , Trans-Activators/antagonists & inhibitors , Trans-Activators/physiology
19.
J Virol ; 82(24): 12392-405, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18842706

ABSTRACT

Coronaviruses express two very large replicase polyproteins, the 16 autoproteolytic cleavage products of which collectively form the membrane-anchored replication complexes. How these structures are assembled is still largely unknown, but it is likely that the membrane-spanning members of these nonstructural proteins (nsps) are responsible for the induction of the double-membrane vesicles and for anchoring the replication complexes to these membranes. For 3 of the 16 coronavirus nsps-nsp3, nsp4, and nsp6-multiple transmembrane domains are predicted. Previously we showed that, consistent with predictions, nsp4 occurs in membranes with both of its termini exposed in the cytoplasm (M. Oostra et al., J. Virol. 81:12323-12336, 2007). Strikingly, however, for both nsp3 and nsp6, predictions based on a multiple alignment of 27 coronavirus genome sequences indicate an uneven number of transmembrane domains. As a consequence, the proteinase domains present in nsp3 and nsp5 would be separated from their target sequences by the lipid bilayer. To look into this incongruity, we studied the membrane disposition of nsp3 and nsp6 of the severe acute respiratory syndrome coronavirus and murine hepatitis virus by analyzing tagged forms of the proteins expressed in cultured cells. Contrary to the predictions, in both viruses, both proteins had their amino terminus, as well as their carboxy terminus, exposed in the cytoplasm. We established that two of the three hydrophobic domains in nsp3 and six of the seven in nsp6 are membrane spanning. Subsequently, we verified that in nsp4, all four hydrophobic domains span the lipid bilayer. The occurrence of conserved non-membrane-spanning hydrophobic domains in nsp3 and nsp6 suggests an important function for these domains in coronavirus replication.


Subject(s)
Cell Membrane/metabolism , Coronavirus/metabolism , Glycoproteins/metabolism , Hydrophobic and Hydrophilic Interactions , Viral Nonstructural Proteins/metabolism , Virus Internalization , Virus Replication , Cell Line , Coronavirus/genetics , Gene Deletion , Glycoproteins/genetics , Mutation/genetics , Protein Binding , Viral Nonstructural Proteins/genetics
20.
Mol Immunol ; 114: 524-534, 2019 10.
Article in English | MEDLINE | ID: mdl-31518856

ABSTRACT

The chemokine CXCL4 has been implicated in several immune diseases. Exposure of monocyte-derived dendritic cells (moDCs) to CXCL4 potentiates the production of inflammatory cytokines in the presence of TLR3 or TLR7/8 agonists. Here we investigated the transcriptional and post-transcriptional events underlying the augmented inflammatory responses in CXCL4-moDCs. Our results indicate that CXCL4-moDCs display an increased expression and secretion of IL-12, IL-23, IL-6 and TNF upon TLR3 activation. Analysis of the cytokine transcripts for the presence of AU-rich elements (ARE), motifs necessary for ARE-mediated mRNA decay, revealed that all these cytokine transcripts are, at least in silico, possibly regulated at the level of mRNA stability. In vitro assays confirmed that mRNA stability of IL6 and TNF, but not IL12B and IL23A, is increased in CXCL4-moDCs. We next screened the expression of ARE-binding proteins (ARE-BPs) and found that TLR stimulation of CXCL4-moDCs induced tristetraprolin (TTP or ZFP36). Increased TTP mRNA expression was found to be a consequence of TTP phospho-mediated inactivation, which over time causes the protein to degrade its own mRNA. Concomitantly with TTP inactivation, we observed increased MAPK p38 signalling, upstream of TTP, in stimulated CXCL4-moDCs. P38 inhibition restored TTP activation and subsequently reduced the production of inflammatory cytokines. Finally, TTP knockdown in moDCs resulted in an increased production of IL6 and TNF after TLR stimulation. Overall, our study shows that the pro-inflammatory phenotype of CXCL4-moDCs relies in part on enhanced cytokine mRNA stability dictated by TTP inactivation.


Subject(s)
Cytokines/metabolism , Dendritic Cells/metabolism , Monocytes/metabolism , Platelet Factor 4/metabolism , RNA, Messenger/metabolism , Humans , Interleukin-12/metabolism , MAP Kinase Signaling System/physiology , RNA Stability/physiology , Signal Transduction/physiology , Tristetraprolin/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL