Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Autoimmun ; 146: 103219, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38696927

ABSTRACT

Tissue repair is disturbed in fibrotic diseases like systemic sclerosis (SSc), where the deposition of large amounts of extracellular matrix components such as collagen interferes with organ function. LAIR-1 is an inhibitory collagen receptor highly expressed on tissue immune cells. We questioned whether in SSc, impaired LAIR-1-collagen interaction is contributing to the ongoing inflammation and fibrosis. We found that SSc patients do not have an intrinsic defect in LAIR-1 expression or function. Instead, fibroblasts from healthy controls and SSc patients stimulated by soluble factors that drive inflammation and fibrosis in SSc deposit disorganized collagen products in vitro, which are dysfunctional LAIR-1 ligands. This is dependent of matrix metalloproteinases and platelet-derived growth factor receptor signaling. In support of a non-redundant role of LAIR-1 in the control of fibrosis, we found that LAIR-1-deficient mice have increased skin fibrosis in response to repeated injury and in the bleomycin mouse model for SSc. Thus, LAIR-1 represents an essential control mechanism for tissue repair. In fibrotic disease, excessive collagen degradation may lead to a disturbed feedback loop. The presence of functional LAIR-1 in patients provides a therapeutic opportunity to reactivate this intrinsic negative feedback mechanism in fibrotic diseases.


Subject(s)
Collagen , Disease Models, Animal , Fibroblasts , Fibrosis , Mice, Knockout , Receptors, Immunologic , Scleroderma, Systemic , Animals , Humans , Scleroderma, Systemic/immunology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Mice , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Collagen/metabolism , Fibroblasts/metabolism , Bleomycin/adverse effects , Skin/pathology , Skin/metabolism , Skin/immunology , Signal Transduction , Male , Female , Cells, Cultured
2.
Cancer Immunol Immunother ; 72(12): 4385-4397, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37938368

ABSTRACT

This study examined the composition of the immune microenvironment at different sites within resected pancreas specimens from patients with pancreatic ductal adenocarcinoma (PDAC). Therefore, single-cell suspensions were made from fresh tumor and non-tumorous tissue. Fourteen patients were included from whom twelve PDAC and five non-tumorous samples were obtained. These samples were analyzed with a nineteen marker panel on the Aurora spectral flow cytometer. Furthermore, slides from formalin-fixed paraffine PDACs of eight additional patients were stained with eight markers and analyzed by multispectral imaging. These corresponded to central tumor, periphery of the tumor, i.e., invasive front and resected lymph node and were divided into tumor and adjacent tissue. In the single-cell suspension, a decreased ratio between lymphoid and myeloid cells and between M1 and M2 macrophages was observed in the tumor tissue compared to non-tumorous tissue. Furthermore, an increase in CD169 + macrophages in patients undergoing neoadjuvant therapy was found. Using immunofluorescence, more macrophages compared to T cells were observed, as well as a lower ratio of CD8 to M2 macrophage, a higher ratio of CD4-CD8 T cells and a higher ratio of immune-suppressive cells to pro-inflammatory cells in the PDAC area compared to the adjacent non-tumorous tissue. Finally, there were more immune-suppressive cells in the central tumor area compared to the invasive front. In conclusion, we show a gradient in the immune-suppressive environment in PDAC from most suppressive in the central tumor to least suppressive in distant non-tumorous tissue.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Tumor Microenvironment , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Pancreas/pathology , T-Lymphocytes
3.
Proc Natl Acad Sci U S A ; 117(44): 27528-27539, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33067394

ABSTRACT

Priming of CD8+ T cells by dendritic cells (DCs) is crucial for the generation of effective antitumor immune responses. Here, we describe a liposomal vaccine carrier that delivers tumor antigens to human CD169/Siglec-1+ antigen-presenting cells using gangliosides as targeting ligands. Ganglioside-liposomes specifically bound to CD169 and were internalized by in vitro-generated monocyte-derived DCs (moDCs) and macrophages and by ex vivo-isolated splenic macrophages in a CD169-dependent manner. In blood, high-dimensional reduction analysis revealed that ganglioside-liposomes specifically targeted CD14+ CD169+ monocytes and Axl+ CD169+ DCs. Liposomal codelivery of tumor antigen and Toll-like receptor ligand to CD169+ moDCs and Axl+ CD169+ DCs led to cytokine production and robust cross-presentation and activation of tumor antigen-specific CD8+ T cells. Finally, Axl+ CD169+ DCs were present in cancer patients and efficiently captured ganglioside-liposomes. Our findings demonstrate a nanovaccine platform targeting CD169+ DCs to drive antitumor T cell responses.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Macrophages/immunology , Neoplasms/therapy , Vaccination/methods , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Cross-Priming/immunology , Dendritic Cells/metabolism , Gangliosides , Humans , Immunogenicity, Vaccine , Leukocytes, Mononuclear , Liposomes , Macrophages/metabolism , Neoplasms/immunology , Primary Cell Culture , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Sialic Acid Binding Ig-like Lectin 1/metabolism , THP-1 Cells , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Axl Receptor Tyrosine Kinase
4.
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
5.
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
6.
Eur J Immunol ; 48(3): 522-531, 2018 03.
Article in English | MEDLINE | ID: mdl-29193036

ABSTRACT

CXCL4 regulates multiple facets of the immune response and is highly upregulated in various Th17-associated rheumatic diseases. However, whether CXCL4 plays a direct role in the induction of IL-17 production by human CD4+ T cells is currently unclear. Here, we demonstrated that CXCL4 induced human CD4+ T cells to secrete IL-17 that co-expressed IFN-γ and IL-22, and differentiated naïve CD4+ T cells to become Th17-cytokine producing cells. In a co-culture system of human CD4+ T cells with monocytes or myeloid dendritic cells, CXCL4 induced IL-17 production upon triggering by superantigen. Moreover, when monocyte-derived dendritic cells were differentiated in the presence of CXCL4, they orchestrated increased levels of IL-17, IFN-γ, and proliferation by CD4+ T cells. Furthermore, the CXCL4 levels in synovial fluid from psoriatic arthritis patients strongly correlated with IL-17 and IL-22 levels. A similar response to CXCL4 of enhanced IL-17 production by CD4+ T cells was also observed in patients with psoriatic arthritis. Altogether, we demonstrate that CXCL4 boosts pro-inflammatory cytokine production especially IL-17 by human CD4+ T cells, either by acting directly or indirectly via myeloid antigen presenting cells, implicating a role for CXCL4 in PsA pathology.


Subject(s)
Arthritis, Psoriatic/immunology , Interleukin-17/biosynthesis , Interleukins/metabolism , Platelet Factor 4/immunology , Th17 Cells/immunology , Antigen-Presenting Cells/immunology , Case-Control Studies , Cell Differentiation/immunology , Coculture Techniques , Dendritic Cells/immunology , Humans , Lymphocyte Activation , Monocytes/immunology , Interleukin-22
7.
Ann Rheum Dis ; 78(9): 1249-1259, 2019 09.
Article in English | MEDLINE | ID: mdl-31126957

ABSTRACT

OBJECTIVES: Systemic sclerosis (SSc) is an autoimmune disease with unknown pathogenesis manifested by inflammation, vasculopathy and fibrosis in skin and internal organs. Type I interferon signature found in SSc propelled us to study plasmacytoid dendritic cells (pDCs) in this disease. We aimed to identify candidate pathways underlying pDC aberrancies in SSc and to validate its function on pDC biology. METHODS: In total, 1193 patients with SSc were compared with 1387 healthy donors and 8 patients with localised scleroderma. PCR-based transcription factor profiling and methylation status analyses, single nucleotide polymorphism genotyping by sequencing and flow cytometry analysis were performed in pDCs isolated from the circulation of healthy controls or patients with SSc. pDCs were also cultured under hypoxia, inhibitors of methylation and hypoxia-inducible factors and runt-related transcription factor 3 (RUNX3) levels were determined. To study Runx3 function, Itgax-Cre:Runx3f/f mice were used in in vitro functional assay and bleomycin-induced SSc skin inflammation and fibrosis model. RESULTS: Here, we show downregulation of transcription factor RUNX3 in SSc pDCs. A higher methylation status of the RUNX3 gene, which is associated with polymorphism rs6672420, correlates with lower RUNX3 expression and SSc susceptibility. Hypoxia is another factor that decreases RUNX3 level in pDC. Mouse pDCs deficient of Runx3 show enhanced maturation markers on CpG stimulation. In vivo, deletion of Runx3 in dendritic cell leads to spontaneous induction of skin fibrosis in untreated mice and increased severity of bleomycin-induced skin fibrosis. CONCLUSIONS: We show at least two pathways potentially causing low RUNX3 level in SSc pDCs, and we demonstrate the detrimental effect of loss of Runx3 in SSc model further underscoring the role of pDCs in this disease.


Subject(s)
Core Binding Factor Alpha 3 Subunit/genetics , Dendritic Cells/metabolism , Gene Expression Regulation , RNA/genetics , Scleroderma, Systemic/genetics , Skin/pathology , Animals , Core Binding Factor Alpha 3 Subunit/biosynthesis , Dendritic Cells/pathology , Disease Models, Animal , Disease Progression , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Humans , Mice , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Skin/metabolism
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 Immunol ; 199(1): 253-262, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28515281

ABSTRACT

Chemokines have been shown to play immune-modulatory functions unrelated to steering cell migration. CXCL4 is a chemokine abundantly produced by activated platelets and immune cells. Increased levels of circulating CXCL4 are associated with immune-mediated conditions, including systemic sclerosis. Considering the central role of dendritic cells (DCs) in immune activation, in this article we addressed the effect of CXCL4 on the phenotype and function of monocyte-derived DCs (moDCs). To this end, we compared innate and adaptive immune responses of moDCs with those that were differentiated in the presence of CXCL4. Already prior to TLR- or Ag-specific stimulation, CXCL4-moDCs displayed a more matured phenotype. We found that CXCL4 exposure can sensitize moDCs for TLR-ligand responsiveness, as illustrated by a dramatic upregulation of CD83, CD86, and MHC class I in response to TLR3 and TLR7/8-agonists. Also, we observed a markedly increased secretion of IL-12 and TNF-α by CXCL4-moDCs exclusively upon stimulation with polyinosinic-polycytidylic acid, R848, and CL075 ligands. Next, we analyzed the effect of CXCL4 in modulating DC-mediated T cell activation. CXCL4-moDCs strongly potentiated proliferation of autologous CD4+ T cells and CD8+ T cells and production of IFN-γ and IL-4, in an Ag-independent manner. Although the internalization of Ag was comparable to that of moDCs, Ag processing by CXCL4-moDCs was impaired. Yet, these cells were more potent at stimulating Ag-specific CD8+ T cell responses. Together our data support that increased levels of circulating CXCL4 may contribute to immune dysregulation through the modulation of DC differentiation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation , Dendritic Cells/immunology , Lymphocyte Activation , Platelet Factor 4/immunology , Tumor Necrosis Factor-alpha/immunology , Antigens, CD/genetics , Antigens, CD/immunology , B7-2 Antigen/genetics , B7-2 Antigen/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/physiology , Cells, Cultured , Coculture Techniques , Dendritic Cells/drug effects , Dendritic Cells/physiology , Escherichia coli/physiology , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/prevention & control , Genes, MHC Class I , Humans , Imidazoles/pharmacology , Immunoglobulins/genetics , Immunoglobulins/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Phenotype , Platelet Factor 4/metabolism , Platelet Factor 4/pharmacology , Poly I-C/pharmacology , Quinolines/pharmacology , Thiazoles/pharmacology , CD83 Antigen
10.
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
11.
N Engl J Med ; 370(5): 433-43, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24350901

ABSTRACT

BACKGROUND: Plasmacytoid dendritic cells have been implicated in the pathogenesis of systemic sclerosis through mechanisms beyond the previously suggested production of type I interferon. METHODS: We isolated plasmacytoid dendritic cells from healthy persons and from patients with systemic sclerosis who had distinct clinical phenotypes. We then performed proteome-wide analysis and validated these observations in five large cohorts of patients with systemic sclerosis. Next, we compared the results with those in patients with systemic lupus erythematosus, ankylosing spondylitis, and hepatic fibrosis. We correlated plasma levels of CXCL4 protein with features of systemic sclerosis and studied the direct effects of CXCL4 in vitro and in vivo. RESULTS: Proteome-wide analysis and validation showed that CXCL4 is the predominant protein secreted by plasmacytoid dendritic cells in systemic sclerosis, both in circulation and in skin. The mean (±SD) level of CXCL4 in patients with systemic sclerosis was 25,624±2652 pg per milliliter, which was significantly higher than the level in controls (92.5±77.9 pg per milliliter) and than the level in patients with systemic lupus erythematosus (1346±1011 pg per milliliter), ankylosing spondylitis (1368±1162 pg per milliliter), or liver fibrosis (1668±1263 pg per milliliter). CXCL4 levels correlated with skin and lung fibrosis and with pulmonary arterial hypertension. Among chemokines, only CXCL4 predicted the risk and progression of systemic sclerosis. In vitro, CXCL4 down-regulated expression of transcription factor FLI1, induced markers of endothelial-cell activation, and potentiated responses of toll-like receptors. In vivo, CXCL4 induced the influx of inflammatory cells and skin transcriptome changes, as in systemic sclerosis. CONCLUSIONS: Levels of CXCL4 were elevated in patients with systemic sclerosis and correlated with the presence and progression of complications, such as lung fibrosis and pulmonary arterial hypertension. (Funded by the Dutch Arthritis Association and others.).


Subject(s)
Dendritic Cells/metabolism , Platelet Factor 4/blood , Scleroderma, Systemic/blood , Adult , Animals , Biomarkers/blood , Cytokines/metabolism , Familial Primary Pulmonary Hypertension , Female , Humans , Hypertension, Pulmonary/blood , Male , Mice , Mice, Inbred C57BL , Middle Aged , Platelet Factor 4/metabolism , Proteome , Pulmonary Fibrosis/blood , RNA, Messenger/metabolism , Scleroderma, Systemic/etiology , Skin/pathology
12.
Exp Dermatol ; 24(2): 133-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25425469

ABSTRACT

Toll-like receptor 9 (TLR9) drives innate immune responses after recognition of foreign or endogenous DNA containing unmethylated CpG motifs. DNA-mediated TLR9 activation is highly implicated in the pathogenesis of several autoimmune skin diseases, yet its contribution to the inflammation seen in these diseases remains unclear. In this study, TLR9 ligand, CpGB DNA, was administered to mice via a subcutaneous osmotic pump with treatment lasting 1 or 4 weeks. Gene expression and immunofluorescence analyses were used to determine chemokine expression and cell recruitment in the skin surrounding the pump outlet. CpGB DNA skin treatment dramatically induced a marked influx of CD11b+ F4/80+ macrophages, increasing over 4 weeks of treatment, and induction of IFNγ and TNFα expression. Chemokines, CCL2, CCL4, CCL5, CXCL9 and CXCL10, were highly induced in CpGB DNA-treated skin, although abrogation of these signalling pathways individually did not alter macrophage accumulation. Flow cytometry analysis showed that TLR9 activation in the skin increased circulating CD11b+ CD115+ Ly6C(hi) inflammatory monocytes following 1 week of CpGB DNA treatment. Additionally, skin-resident CD11b+ cells were found to initially take up subcutaneous CpGB DNA and propagate the subsequent immune response. Using diphtheria toxin-induced monocyte depletion mouse model, gene expression analysis demonstrated that CD11b+ cells are responsible for the CpGB DNA-induced cytokine and chemokine response. Overall, these data demonstrate that chronic TLR9 activation induces a specific inflammatory response, ultimately leading to a striking and selective accumulation of macrophages in the skin.


Subject(s)
CpG Islands , Macrophage Activation , Macrophages/metabolism , Skin/metabolism , Administration, Cutaneous , Animals , CD11b Antigen/metabolism , Chemokines/metabolism , DNA , Diphtheria Toxin/chemistry , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Immunohistochemistry , Inflammation/metabolism , Interferon-gamma/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Monocytes/cytology , Osmosis , Skin/cytology , Toll-Like Receptor 9/metabolism
13.
Ann Rheum Dis ; 73(10): 1864-72, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23873879

ABSTRACT

OBJECTIVE: To characterise global chemokine expression in systemic sclerosis (SSc) skin in order to better understand the relationship between chemokine expression and vascular inflammation in this disease. METHODS: We investigated chemokine mRNA expression in the skin through quantitative PCR analysis comparing patients with diffuse cutaneous (dcSSc) or limited cutaneous (lcSSc) disease with healthy controls. We tested correlations between the most regulated chemokines and vascular inflammation and macrophage recruitment. CCL19 expression was examined in human primary immune cells treated with innate immune activators. RESULTS: The chemokines, CCL18, CCL19 and CXCL13, were upregulated in dcSSc skin, and CCL18 in lcSSc skin. Expression of CCL19 in dcSSc skin correlated with markers of vascular inflammation and macrophage recruitment. Immunofluorescence data showed CCL19 colocalisation with CD163 macrophages in dcSSc skin. In vitro studies on human primary cells demonstrated that CCL19 expression was induced after toll-like receptor activation of peripheral blood mononuclear cells and separated populations of CD14 monocytes. CONCLUSIONS: CCL18, CCL19 and CXCL13-chemoattractants for macrophage and T cell recruitment-were three of six chemokines with the highest expression in dcSSc skin. Increased CCL19 expression in the skin suggests a role for CCL19 in the recruitment of immune cells to the peripheral tissue. Induction of CCL19 in macrophages but not structural cells indicates a role for skin-resident or recruited immune cells in perivascular inflammation. This study demonstrates that CCL19 is a sensitive marker for the perivascular inflammation and immune cell recruitment seen in dcSSc skin disease.


Subject(s)
Chemokines/biosynthesis , Scleroderma, Systemic/immunology , Skin Diseases, Vascular/immunology , Vasculitis/immunology , Adult , Aged , Biomarkers/metabolism , Case-Control Studies , Cells, Cultured , Chemokine CCL19/biosynthesis , Chemokine CCL19/genetics , Chemokines/genetics , Female , Gene Expression/immunology , Gene Expression Profiling/methods , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Leukocytes, Mononuclear/immunology , Macrophage Activation/genetics , Macrophage Activation/immunology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/genetics , Scleroderma, Systemic/complications , Scleroderma, Systemic/genetics , Skin/immunology , Skin Diseases, Vascular/etiology , Skin Diseases, Vascular/genetics , Up-Regulation/immunology , Vasculitis/etiology , Vasculitis/genetics
14.
Arthritis Rheum ; 65(5): 1335-46, 2013 May.
Article in English | MEDLINE | ID: mdl-23335246

ABSTRACT

OBJECTIVE: To explore the expression of thymic stromal lymphopoietin (TSLP) in patients with diffuse cutaneous systemic sclerosis (dcSSc) and compare its effects in vivo and in vitro with those of interleukin-13 (IL-13) and transforming growth factor ß (TGFß). METHODS: Skin biopsy specimens from patients with dcSSc (n = 14) and healthy controls (n = 13) were analyzed by immunohistochemistry and immunofluorescence for TSLP, TSLP receptor, CD4, CD8, CD31, and CD163 markers. Wild-type, IL-4Rα1-, and TSLP-deficient mice were treated with TGFß, IL-13, poly(I-C), or TSLP by osmotic pump. Human fibroblasts and peripheral blood mononuclear cells (PBMCs) were stimulated with TGFß, IL-13, poly(I-C), or TSLP. Microarray analysis and quantitative polymerase chain reaction were performed to determine gene expression, and protein levels of phospho-Smad2 and macrophage marker CD163 were tested. RESULTS: TSLP was highly expressed in the skin of dcSSc patients, more strongly in perivascular areas and in immune cells, and was produced mainly by CD163+ cells. The skin of TSLP-treated mice showed up-regulated clusters of gene expression that overlapped strongly with those in IL-13- and TGFß-treated mice. TSLP up-regulated specific genes, including CXCL9, proteasome, and interferon (IFN)-regulated genes. TSLP treatment in IL-4Rα1-deficient mice promoted similar cutaneous inflammation as in wild-type mice, though TSLP-induced arginase 1, CCL2, and matrix metalloproteinase 12 messenger RNA levels were blocked. In PBMCs, TSLP up-regulated tumor necrosis factor α, Mx-1, IFNγ, CXCL9, and mannose receptor 1 gene expression. TSLP-deficient mice treated with TGFß showed less fibrosis and blocked expression of plasminogen activator inhibitor 1 and osteopontin 1. Poly(I-C)-treated mice showed high levels of cutaneous TSLP. CONCLUSION: TSLP is highly expressed in the skin of dcSSc patients and interacts in a complex manner with 2 other profibrotic cytokines, TGFß and IL-13, strongly suggesting that it might promote SSc fibrosis directly or indirectly by synergistically stimulating profibrotic genes, or production of these cytokines.


Subject(s)
Cytokines/biosynthesis , Fibrosis/genetics , Interleukin-13/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Scleroderma, Diffuse/metabolism , Skin/metabolism , Transforming Growth Factor beta/metabolism , Animals , Biomarkers/metabolism , Cytokines/deficiency , Cytokines/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression/drug effects , Humans , Interleukin-13/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Poly I-C/pharmacology , RNA, Messenger/metabolism , Scleroderma, Diffuse/diagnosis , Skin/pathology , Transforming Growth Factor beta/pharmacology , Up-Regulation , Thymic Stromal Lymphopoietin
15.
Tissue Eng Regen Med ; 21(3): 455-471, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38114886

ABSTRACT

BACKGROUND: Human lymph node (HuLN) models have emerged with invaluable potential for immunological research and therapeutic application given their fundamental role in human health and disease. While fibroblastic reticular cells (FRCs) are instrumental to HuLN functioning, their inclusion and recognition of importance for organotypic in vitro lymphoid models remain limited. METHODS: Here, we established an in vitro three-dimensional (3D) model in a collagen-fibrin hydrogel with primary FRCs and a dendritic cell (DC) cell line (MUTZ-3 DC). To study and characterise the cellular interactions seen in this 3D FRC-DC organotypic model compared to the native HuLN; flow cytometry, immunohistochemistry, immunofluorescence and cytokine/chemokine analysis were performed. RESULTS: FRCs were pivotal for survival, proliferation and localisation of MUTZ-3 DCs. Additionally, we found that CD1a expression was absent on MUTZ-3 DCs that developed in the presence of FRCs during cytokine-induced MUTZ-3 DC differentiation, which was also seen with primary monocyte-derived DCs (moDCs). This phenotype resembled HuLN-resident DCs, which we detected in primary HuLNs, and these CD1a- MUTZ-3 DCs induced T cell proliferation within a mixed leukocyte reaction (MLR), indicating a functional DC status. FRCs expressed podoplanin (PDPN), CD90 (Thy-1), CD146 (MCAM) and Gremlin-1, thereby resembling the DC supporting stromal cell subset identified in HuLNs. CONCLUSION: This 3D FRC-DC organotypic model highlights the influence and importance of FRCs for DC functioning in a more realistic HuLN microenvironment. As such, this work provides a starting point for the development of an in vitro HuLN.


Subject(s)
Cytokines , Microphysiological Systems , Humans , Cell Line , Cytokines/metabolism , Dendritic Cells/metabolism , Lymph Nodes/metabolism
16.
Front Immunol ; 14: 1290272, 2023.
Article in English | MEDLINE | ID: mdl-38054006

ABSTRACT

Historically platelets are mostly known for their crucial contribution to hemostasis, but there is growing understanding of their role in inflammation and immunity. The immunomodulatory role of platelets entails interaction with pathogens, but also with immune cells including macrophages and dendritic cells (DCs), to activate adaptive immune responses. In our previous work, we have demonstrated that splenic CD169+ macrophages scavenge liposomes and collaborate with conventional type 1 DCs (cDC1) to induce expansion of CD8+ T cells. Here, we show that platelets associate with liposomes and bind to DNGR-1/Clec9a and CD169/Siglec-1 receptors in vitro. In addition, platelets interacted with splenic CD169+ macrophages and cDC1 and further increased liposome internalization by cDC1. Most importantly, platelet depletion prior to liposomal immunization resulted in significantly diminished antigen-specific CD8+ T cell responses, but not germinal center B cell responses. Previously, complement C3 was shown to be essential for platelet-mediated CD8+ T cell activation during bacterial infection. However, after liposomal vaccination CD8+ T cell priming was not dependent on complement C3. While DCs from platelet-deficient mice exhibited unaltered maturation status, they did express lower levels of CCR7. In addition, in the absence of platelets, CCL5 plasma levels were significantly reduced. Overall, our findings demonstrate that platelets engage in a cross-talk with CD169+ macrophages and cDC1 and emphasize the importance of platelets in induction of CD8+ T cell responses in the context of liposomal vaccination.


Subject(s)
CD8-Positive T-Lymphocytes , Liposomes , Animals , Mice , Liposomes/metabolism , Complement C3/metabolism , Macrophages , Antigens
17.
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
18.
Arthritis Rheum ; 63(6): 1718-28, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21425123

ABSTRACT

OBJECTIVE: To explore the relationship between biomarkers of pulmonary arterial hypertension (PAH), interferon (IFN)-regulated gene expression, and the alternative activation pathway in systemic sclerosis (SSc). METHODS: Peripheral blood mononuclear cells (PBMCs) were purified from healthy controls, patients with idiopathic PAH, and SSc patients (classified as having diffuse cutaneous SSc, limited cutaneous SSc [lcSSc] without PAH, and lcSSc with PAH). IFN-regulated and "PAH biomarker" genes were compared after supervised hierarchical clustering. Messenger RNA levels of selected IFN-regulated genes (Siglec1 and MX1), biomarker genes (IL13RA1, CCR1, and JAK2), and the alternative activation marker gene (MRC1) were analyzed on PBMCs and on CD14- and CD14+ cell populations. Interleukin-13 (IL-13) and IL-4 concentrations were measured in plasma by immunoassay. CD14, MRC1, and IL13RA1 surface expression was analyzed by flow cytometry. RESULTS: Increased PBMC expression of both IFN-regulated and biomarker genes distinguished SSc patients from healthy controls. Expression of genes in the biomarker cluster, but not in the IFN-regulated cluster, distinguished lcSSc with PAH from lcSSc without PAH. The genes CCR1 (P<0.001) and JAK2 (P<0.001) were expressed more highly in lcSSc patients with PAH compared with controls and mainly by CD14+ cells. MRC1 expression was increased exclusively in lcSSc patients with PAH (P<0.001) and correlated strongly with pulmonary artery pressure (r=0.52, P=0.03) and higher mortality (P=0.02). MRC1 expression was higher in CD14+ cells and was greatly increased by stimulation with IL-13. IL-13 concentrations in plasma were most highly increased in lcSSc patients with PAH (P<0.001). CONCLUSION: IFN-regulated and biomarker genes represent distinct, although related, clusters in lcSSc patients with PAH. MRC1, a marker for the effect of IL-13 on alternative monocyte/macrophage activation, is associated with this severe complication and is related to mortality.


Subject(s)
Interferons/immunology , Macrophage Activation/immunology , Scleroderma, Systemic/immunology , Adult , Aged , Familial Primary Pulmonary Hypertension , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Gene Expression Regulation , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/mortality , Interleukin-13/blood , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/immunology , Interleukin-4/blood , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Lipopolysaccharide Receptors/immunology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Middle Aged , Myxovirus Resistance Proteins , Receptors, CCR1/genetics , Receptors, CCR1/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Scleroderma, Systemic/complications , Sialic Acid Binding Ig-like Lectin 1
19.
Front Immunol ; 13: 842241, 2022.
Article in English | MEDLINE | ID: mdl-35251040

ABSTRACT

Cancer vaccination aims to activate immunity towards cancer cells and can be achieved by delivery of cancer antigens together with immune stimulatory adjuvants to antigen presenting cells (APC). APC maturation and antigen processing is a subsequent prerequisite for T cell priming and anti-tumor immunity. In order to specifically target APC, nanoparticles, such as liposomes, can be used for the delivery of antigen and adjuvant. We have previously shown that liposomal inclusion of the ganglioside GM3, an endogenous ligand for CD169, led to robust uptake by CD169-expressing APC and resulted in strong immune responses when supplemented with a soluble adjuvant. To minimize the adverse effects related to a soluble adjuvant, immune stimulatory molecules can be incorporated in liposomes to achieve targeted delivery of both antigen and adjuvant. In this study, we incorporated TLR4 (MPLA) or TLR7/8 (3M-052) ligands in combination with inflammasome stimuli, 1-palmitoyl-2-glutaryl-sn-glycero-3-phosphocholine (PGPC) or muramyl dipeptide (MDP), into GM3 liposomes. Incorporation of TLR and inflammasome ligands did not interfere with the uptake of GM3 liposomes by CD169-expressing cells. GM3 liposomes containing a TLR ligand efficiently matured human and mouse dendritic cells in vitro and in vivo, while inclusion of PGPC or MDP had minor effects on maturation. Immunization with MPLA-containing GM3 liposomes containing an immunogenic synthetic long peptide stimulated CD4+ and CD8+ T cell responses, but additional incorporation of either PGPC or MDP did not translate into stronger immune responses. In conclusion, our study indicates that TLRL-containing GM3 liposomes are effective vectors to induce DC maturation and T cell priming and thus provide guidance for further selection of liposomal components to optimally stimulate anti-cancer immune responses.


Subject(s)
Liposomes , Neoplasms , Adjuvants, Immunologic/pharmacology , Animals , Antigens/metabolism , Dendritic Cells , Inflammasomes/metabolism , Ligands , Liposomes/chemistry , Mice , Toll-Like Receptors/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL