Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Immunol ; 209(10): 1906-1917, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36426957

ABSTRACT

Rheumatoid arthritis (RA) is characterized by autoimmune joint destruction with debilitating consequences. Despite treatment advancements with biologic therapies, a significant proportion of RA patients show an inadequate clinical response, and restoration of immune self-tolerance represents an unmet therapeutic need. We have previously described a tolerogenic phenotype of plasmacytoid dendritic cells (pDCs) in RA patients responding to anti-TNF-α agents. However, the molecular mechanisms involved in tolerogenic reprogramming of pDCs in RA remain elusive. In this study, guided by transcriptomic analysis of CD303+CD123+ pDCs from RA patients in remission, we revealed enhanced expression of IL-6R and its downstream signaling compared with healthy pDCs. Functional assessment demonstrated that IL-6R engagement resulted in marked reduction of TNF-α secretion by pDCs whereas intracellular TNF-α was significantly increased. Accordingly, pharmacologic inhibition of IL-6R signaling restored TNF-α secretion levels by pDCs. Mechanistic analysis demonstrated impaired activity and decreased lysosomal degradation of ADAM17 (a disintegrin and metalloproteinase 17) sheddase in pDCs, which is essential for TNF-α cleavage. Importantly, reduction of TNF-α secretion by IL-6-treated pDCs attenuated the inflammatory potential of RA patient-derived synovial fibroblasts. Collectively, these findings position pDCs as an important source of TNF-α in RA pathogenesis and unravel an anti-inflammatory mechanism of IL-6 by limiting the pDC-derived TNF-α secretion.


Subject(s)
Arthritis, Rheumatoid , Interleukin-6 , Humans , Tumor Necrosis Factor Inhibitors , Dendritic Cells , Signal Transduction , Tumor Necrosis Factor-alpha
2.
Arthritis Res Ther ; 23(1): 246, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34560894

ABSTRACT

BACKGROUND: In patients with axial spondyloarthritis (axSpA), monocytes show a pre-activated phenotype. Gut inflammation is a trigger of monocyte activation and may also affect their development in the bone marrow (BM). As gut inflammation is commonly observed in axSpA patients, we performed a detailed analysis of monocyte transcriptomes of axSpA patients in two cohorts and searched for signs of activation and developmental adaptations as putative imprints of gut inflammation. METHODS: Transcriptomes of blood CD14+ monocytes of HLA-B27+ axSpA patients and healthy controls (HC) were generated by microarrays from cohort 1 and by RNA-sequencing from cohort 2. Differentially expressed genes from both analyses were subjected to gene set enrichment analysis (GSEA) and to co-expression analysis in reference transcriptomes from BM cells, blood cells and activated monocytes. As serological markers of translocation, 1,3 beta-glycan, intestinal fatty acid binding protein, and lipopolysaccharide binding protein (LBP) were determined by LAL and ELISA. RESULTS: Transcriptome analysis identified axSpA-specific monocyte signatures showing an imprint of LPS/cytokine-activated monocytes, late granulopoietic BM cells, blood neutrophils, and G-CSF-mobilized blood cells, which suggests LPS/TNF activation and more prominent BM adaptation promoting a neutrophil-like phenotype. GSEA mapped axSpA upregulated genes to inflammatory responses and TNFα signaling and downregulated probe-sets to metabolic pathways. Among translocation markers, LBP levels were significantly increased in axSpA patients vs. HC (p < 0.001). Stratified analysis by disease activity and stage identified an "active disease signature" (BASDAI ≥ 4) with an imprint of LPS/cytokine-activated monocytes and CD16+ monocyte subsets. The "AS signature" (vs. non-radiographic axSpA) showed a reinforced neutrophil-like phenotype due to deprivation of dendritic cell transcripts. CONCLUSIONS: The neutrophil-like phenotype of axSpA monocytes points towards a biased monocytopoiesis from granulocyte-monocyte progenitors. This shift in monocytopoiesis and the LPS/cytokine imprint as well as the elevated LBP levels are indicators of systemic inflammation, which may result from bacterial translocation. The BM adaptation is most prominent in AS patients while disease activity appears to be linked to activation and trafficking of monocytes.


Subject(s)
Monocytes , Spondylarthritis , Cytokines , Gene Expression Profiling , Humans , Spondylarthritis/genetics , Transcriptome
3.
Sci Rep ; 10(1): 7907, 2020 05 13.
Article in English | MEDLINE | ID: mdl-32404914

ABSTRACT

Advances in microbiome research suggest involvement in chronic inflammatory diseases such as rheumatoid arthritis (RA). Searching for initial trigger(s) in RA, we compared transcriptome profiles of highly inflamed RA synovial tissue (RA-ST) and osteoarthritis (OA)-ST with 182 selected reference transcriptomes of defined cell types and their activation by exogenous (microbial) and endogenous inflammatory stimuli. Screening for dominant changes in RA-ST demonstrated activation of monocytes/macrophages with gene-patterns induced by bacterial and fungal triggers. Gene-patterns of activated B- or T-cells in RA-ST reflected a response to activated monocytes/macrophages rather than inducing their activation. In contrast, OA-ST was dominated by gene-patterns of non-activated macrophages and fibroblasts. The difference between RA and OA was more prominent in transcripts of secreted proteins and was confirmed by protein quantification in synovial fluid (SF) and serum. In total, 24 proteins of activated cells were confirmed in RA-SF compared to OA-SF and some like CXCL13, CCL18, S100A8/A9, sCD14, LBP reflected this increase even in RA serum. Consequently, pathogen-like response patterns in RA suggest that direct microbial influences exist. This challenges the current concept of autoimmunity and immunosuppressive treatment and advocates new diagnostic and therapeutic strategies that consider microbial persistence as important trigger(s) in the etiopathogenesis of RA.


Subject(s)
Arthritis, Rheumatoid/etiology , Arthritis, Rheumatoid/metabolism , Gene Expression Profiling , Macrophage Activation/immunology , Macrophages/immunology , Synovial Membrane/metabolism , Transcriptome , Adaptive Immunity , Arthritis, Rheumatoid/pathology , Biomarkers , Disease Susceptibility , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Macrophages/metabolism , Monocytes/immunology , Monocytes/metabolism , Organ Specificity , Severity of Illness Index
4.
Arthritis Rheumatol ; 71(10): 1756-1765, 2019 10.
Article in English | MEDLINE | ID: mdl-31131995

ABSTRACT

OBJECTIVE: Inflamed tissue is characterized by low availability of oxygen and nutrients. Yet CD4+ T helper lymphocytes persist over time in such tissue and probably contribute to the chronicity of inflammation. This study was undertaken to analyze the metabolic adaptation of these cells to the inflamed environment. METHODS: Synovial and blood CD4+ T cells isolated ex vivo from patients with juvenile idiopathic arthritis (JIA) and murine CD4+ T cells were either stimulated once or stimulated repeatedly. Their dependency on particular metabolic pathways for survival was then analyzed using pharmacologic inhibitors. The role of the transcription factor Twist 1 was investigated by determining lactate production and oxygen consumption in Twist1-sufficient and Twist1-deficient murine T cells. The dependency of these murine cells on particular metabolic pathways was analyzed using pharmacologic inhibitors. RESULTS: Programmed death 1 (PD-1)+ T helper cells in synovial fluid samples from patients with JIA survived via fatty acid oxidation (mean ± SEM survival of 3.4 ± 2.85% in the presence of etomoxir versus 60 ± 7.08% in the absence of etomoxir on day 4 of culture) (P < 0.0002; n = 6) and expressed the E-box-binding transcription factor TWIST1 (2-14-fold increased expression) (P = 0.0156 versus PD-1- T helper cells; n = 6). Repeatedly restimulated murine T helper cells, which expressed Twist1 as well, needed Twist1 to survive via fatty acid oxidation. In addition, Twist1 protected the cells against reactive oxygen species. CONCLUSION: Our findings indicate that TWIST1 is a master regulator of metabolic adaptation of T helper cells to chronic inflammation and a target for their selective therapeutic elimination.


Subject(s)
Arthritis, Juvenile/metabolism , Fatty Acids/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Twist-Related Protein 1/genetics , Animals , Arthritis, Juvenile/immunology , Cell Survival , Energy Metabolism , Glycolysis , Humans , Inflammation , Lactic Acid/metabolism , Mice , Nuclear Proteins/genetics , Oxidation-Reduction , Oxygen Consumption , Programmed Cell Death 1 Receptor/metabolism , Reactive Oxygen Species/metabolism , Synovial Fluid , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Twist-Related Protein 1/metabolism
5.
Arthritis Res Ther ; 20(1): 191, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30157966

ABSTRACT

BACKGROUND: Therapeutic targeting of tumour necrosis factor (TNF)-α is highly effective in ankylosing spondylitis (AS) patients. However, since one-third of anti-TNF-treated AS patients do not show an adequate clinical response there is an urgent need for new biomarkers that would aid clinicians in their decision-making to select appropriate therapeutic options. Thus, the aim of this explorative study was to identify cell-based biomarkers in peripheral blood that could be used for a pre-treatment stratification of AS patients. METHODS: A high-dimensional, multi-parametric flow cytometric approach was applied to identify baseline predictors in 31 AS patients before treatment with the TNF blockers adalimumab (TNF-neutralisation) and etanercept (soluble TNF receptor). RESULTS: As the major result, the frequencies of natural killer (NK) cells, and in particular CD8-positive (CD8+) NK cell subsets, were most predictive for therapeutic outcome in AS patients. While an inverse correlation between classical CD56+/CD16+ NK cells and reduction of disease activity was observed, the CD8+ NK cell subset behaved in the opposite direction. At baseline, responders showed significantly increased frequencies of CD8+ NK cells compared with non-responders. CONCLUSIONS: This is the first study demonstrating that the composition of the NK cell compartment has predictive power for prediction of therapeutic outcome for anti-TNF-α blockers, and we identified CD8+ NK cells as a potential new player in the TNF-α-driven chronic inflammatory immune response of AS.


Subject(s)
Adalimumab/therapeutic use , Etanercept/therapeutic use , Leukocytes, Mononuclear/drug effects , Spondylitis, Ankylosing/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adalimumab/immunology , Adult , Antirheumatic Agents/immunology , Antirheumatic Agents/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Etanercept/immunology , Female , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Prognosis , Spondylitis, Ankylosing/immunology , Spondylitis, Ankylosing/metabolism , Treatment Outcome , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
6.
Ann Rheum Dis ; 77(2): 300-308, 2018 02.
Article in English | MEDLINE | ID: mdl-29191820

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA) accompanies infiltration and activation of monocytes in inflamed joints. We investigated dominant alterations of RA monocytes in bone marrow (BM), blood and inflamed joints. METHODS: CD14+ cells from BM and peripheral blood (PB) of patients with RA and osteoarthritis (OA) were profiled with GeneChip microarrays. Detailed functional analysis was performed with reference transcriptomes of BM precursors, monocyte blood subsets, monocyte activation and mobilisation. Cytometric profiling determined monocyte subsets of CD14++CD16-, CD14++CD16+ and CD14+CD16+ cells in BM, PB and synovial fluid (SF) and ELISAs quantified the release of activation markers into SF and serum. RESULTS: Investigation of genes differentially expressed between RA and OA monocytes with reference transcriptomes revealed gene patterns of early myeloid precursors in RA-BM and late myeloid precursors along with reduced terminal differentiation to CD14+CD16+monocytes in RA-PB. Patterns associated with tumor necrosis factor/lipopolysaccharide (TNF/LPS) stimulation were weak and more pronounced in RA-PB than RA-BM. Cytometric phenotyping of cells in BM, blood and SF disclosed differences related to monocyte subsets and confirmed the reduced frequency of terminally differentiated CD14+CD16+monocytes in RA-PB. Monocyte activation in SF was characterised by the predominance of CD14++CD16++CD163+HLA-DR+ cells and elevated concentrations of sCD14, sCD163 and S100P. CONCLUSION: Patterns of less mature and less differentiated RA-BM and RA-PB monocytes suggest increased turnover with accelerated monocytopoiesis, BM egress and migration into inflamed joints. Predominant activation in the joint indicates the action of local and primary stimuli, which may also promote adaptive immune triggering through monocytes, potentially leading to new diagnostic and therapeutic strategies.


Subject(s)
Arthritis, Rheumatoid/pathology , Bone Marrow/pathology , Joints/pathology , Monocytes/cytology , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling/methods , Humans , Monocytes/metabolism , Monocytes/pathology , Osteoarthritis/genetics , Osteoarthritis/immunology , Osteoarthritis/pathology , Synovial Fluid/cytology
7.
J Cell Physiol ; 232(6): 1326-1336, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27636101

ABSTRACT

Familial Mediterranean fever (FMF) is an autosomal recessive disease characterized by recurrent, acute, and self-limiting attacks of fever. Mutations in MEFV gene encoding pyrin account for FMF, but the high number of heterozygote patients with typical symptoms of the disease has driven a number of alternative aetiopathogenic hypotheses. The MEFV gene was knocked down in human myelomonocytic cells that express endogenous pyrin to identify deregulated microRNAs (miRNAs). Microarray analyses revealed 29 significantly differentially expressed miRNAs implicated in pathways associated with cellular integrity and survival. Implementation of in silico gene network prediction algorithms and bioinformatics analyses showed that miR-4520a is predicted to target genes implicated in autophagy through regulation of RHEB/mTOR signaling. Differential expression levels of RHEB were confirmed by luciferase reporter gene assays providing further evidence that is directly targeted by miR-4520a. Although the relative expression levels of miR-4520a were variable among FMF patients, the statistical expression of miR-4520a was different between FMF mutation carriers and controls (P = 0.0061), indicating an association between miR-4520a expression and MEFV mutations. Comparison between FMF patients bearing the M694V mutation, associated with severe disease, and healthy controls showed a significant increase in miR-4520a expression levels (P = 0.00545). These data suggest that RHEB, the main activator of mTOR signaling, is a valid target of miR-4520a with the relative expression levels of the latter being significantly deregulated in FMF patients and highly dependent on the presence of pyrin mutations, especially of the M694V type. These results suggest a role of deregulated autophagy in the pathogenesis of FMF. J. Cell. Physiol. 232: 1326-1336, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Familial Mediterranean Fever/genetics , Gene Expression Profiling , MicroRNAs/genetics , Mutation/genetics , Pyrin/genetics , Adult , Case-Control Studies , Cell Line , Female , Gene Regulatory Networks , Humans , Luciferases/metabolism , Male , MicroRNAs/metabolism , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ras Homolog Enriched in Brain Protein
8.
Blood ; 128(26): 3125-3136, 2016 12 29.
Article in English | MEDLINE | ID: mdl-27702798

ABSTRACT

Metabolic changes drive monocyte differentiation and fate. Although abnormal mitochondria metabolism and innate immune responses participate in the pathogenesis of many inflammatory disorders, molecular events regulating mitochondrial activity to control life and death in monocytes remain poorly understood. We show here that, in human monocytes, microRNA-125b (miR-125b) attenuates the mitochondrial respiration through the silencing of the BH3-only proapoptotic protein BIK and promotes the elongation of the mitochondrial network through the targeting of the mitochondrial fission process 1 protein MTP18, leading to apoptosis. Proinflammatory activation of monocyte-derived macrophages is associated with a concomitant increase in miR-125b expression and decrease in BIK and MTP18 expression, which lead to reduced oxidative phosphorylation and enhanced mitochondrial fusion. In a chronic inflammatory systemic disorder, CD14+ blood monocytes display reduced miR-125b expression as compared with healthy controls, inversely correlated with BIK and MTP18 messenger RNA expression. Our findings not only identify BIK and MTP18 as novel targets for miR-125b that control mitochondrial metabolism and dynamics, respectively, but also reveal a novel function for miR-125b in regulating metabolic adaptation of monocytes to inflammation. Together, these data unravel new molecular mechanisms for a proapoptotic role of miR-125b in monocytes and identify potential targets for interfering with excessive inflammatory activation of monocytes in inflammatory disorders.


Subject(s)
Inflammation/genetics , Inflammation/pathology , MicroRNAs/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Monocytes/metabolism , Monocytes/pathology , Aged , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Polarity/genetics , Cell Respiration/genetics , Female , Gene Expression Regulation , Gene Silencing , HEK293 Cells , Humans , Lipopolysaccharide Receptors/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Biological , Toll-Like Receptor 4/metabolism
9.
Transplantation ; 100(4): 898-907, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26444957

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs, miR) hold important roles in the posttranscriptional regulation of gene expression. Their function has been correlated with kidney disease, and they might represent a new class of biomarkers for frequent evaluation of renal graft status. We analyzed their potential in identifying severe T cell-mediated vascular rejection (TCMVR) (Banff 4-II/III) in kidney transplanted patients. METHODS: Microarray experiments and semiquantitative real-time reverse transcription polymerase chain reaction were performed with total RNA isolated from blood cells of kidney graft recipients. Initial microarray analysis revealed 23 differentially expressed miRNAs distinguishing patients with TCMVR from patients with stable grafts. From these, we validated and further determined the expression of 6 differentially expressed miRNAs and 2 control miRNAs in 161 samples from patients with T cell-mediated rejection (Banff 3-Borderline, Banff 4-I/II/III), Banff-2 antibody-mediated rejection, Banff-5 interstitial fibrosis/tubular atrophy, in samples from stable patients and in samples from patients with urinary tract infection using real-time reverse transcription polymerase chain reaction. RESULTS: Expression levels of all 6 candidate miRNAs were significantly downregulated in blood of TCMVR patients compared to the other groups and displayed high sensitivities and specificities for diagnosing TCMVR. The combination of 5 miRNAs, identified by an unbiased multivariate logistic regression followed by cross-validation, enhanced the sensitivity and specificity for the diagnosis of TCMVR after renal transplantation. CONCLUSIONS: The combined measurement of miRNA-15B, miRNA-16, miRNA-103A, miRNA-106A, and miRNA-107 may help to better identify TCMVR after renal transplantation in a precise and clinically applicable way.


Subject(s)
Graft Rejection/blood , Graft Rejection/genetics , Immunity, Cellular/genetics , Kidney Transplantation/adverse effects , MicroRNAs/blood , MicroRNAs/genetics , T-Lymphocytes/immunology , Area Under Curve , Cluster Analysis , Down-Regulation , Gene Expression Profiling/methods , Genetic Markers , Graft Rejection/diagnosis , Graft Rejection/immunology , Humans , Logistic Models , Multivariate Analysis , Oligonucleotide Array Sequence Analysis , Predictive Value of Tests , ROC Curve , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Treatment Outcome
10.
Ann Rheum Dis ; 75(2): 413-21, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25414238

ABSTRACT

OBJECTIVE: Arthritis is a chronic inflammatory disease characterised by immune cell infiltration and mesenchymal cell expansion in the joints. Although the role of immune cells in arthritis is well characterised, the development of mesenchymal cell hyperplasia needs to be better defined. Here, we analysed the role of the ribosomal S6 kinase Rsk2, which we found to be highly activated in joints of patients with arthritis, in the development of mesenchymal cell hyperplasia. METHODS: We genetically inactivated Rsk2 in the tumour necrosis factor (TNF)-α transgenic (TNFtg) mice, an animal model for human inflammatory arthritis. Clinical and histological signs of arthritis as well as molecular markers of inflammation and joint destruction were quantified. Fibroblast-like synoviocytes (FLS) were characterised in vitro and the effect of Rsk2 deletion on the pattern of gene expression was determined. RESULTS: Rsk2 deficiency in TNFtg mice results in earlier and exacerbated inflammation as well as increased bone and cartilage destruction. The production of inflammatory cytokines, matrix metalloproteinases and osteoclastogenic molecules was significantly increased in vivo upon Rsk2 inactivation. Bone marrow deficient in Rsk2 could not transfer this phenotype, indicating that Rsk2 expression in mesenchymal cells controls the course of arthritis. Indeed, Rsk2 deficiency was associated with a more activated phenotype and higher proliferative capacity of FLS, thereby increasing cytokines and production of matrix proteinases. CONCLUSIONS: Rsk2 emerges as a key regulator of mesenchymal cell numbers in the joint and thereby could be targeted to control the inflammatory and tissue-destructive feature of joints in arthritis.


Subject(s)
Arthritis, Experimental/pathology , Fibroblasts/pathology , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Synovial Membrane/pathology , Animals , Arthritis, Experimental/metabolism , Cell Proliferation , Cytokines/metabolism , Disease Models, Animal , Fibroblasts/metabolism , Hyperplasia/genetics , Hyperplasia/metabolism , Inflammation/metabolism , Matrix Metalloproteinases/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Ribosomal Protein S6 Kinases, 90-kDa/deficiency , Synovial Membrane/metabolism , Tumor Necrosis Factor-alpha/genetics
11.
J Immunol ; 194(12): 5812-24, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25948818

ABSTRACT

Foxp3(+) regulatory T cell (Treg)-based immunotherapy holds promise for autoimmune diseases. However, this effort has been hampered by major caveats, including the low frequency of autoantigen-specific Foxp3(+) Tregs and lack of understanding of their molecular and cellular targets, in an unmanipulated wild-type (WT) immune repertoire. In this study, we demonstrate that infusion of myelin in WT mice results in the de novo induction of myelin-specific Foxp3(+) Tregs in WT mice and amelioration of experimental autoimmune encephalomyelitis. Myelin-specific Foxp3(+) Tregs exerted their effect both by diminishing Ag-bearing inflammatory dendritic cell (iDC) recruitment to lymph nodes and by impairing their function. Transcriptome analysis of ex vivo-isolated Treg-exposed iDCs showed significant enrichment of transcripts involved in functional properties of iDCs, including chemotaxis-related genes. To this end, CCR7 expression by iDCs was significantly downregulated in tolerant mice and this was tightly regulated by the presence of IL-10. Collectively, our data demonstrate a novel model for deciphering the Ag-specific Foxp3(+) Treg-mediated mechanisms of tolerance and delineate iDCs as a Foxp3(+) Treg cellular target in unmanipulated mice.


Subject(s)
Autoantigens/immunology , Dendritic Cells/immunology , Inflammation/immunology , Lymph Nodes/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity , Chemotaxis/immunology , Cluster Analysis , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Immune Tolerance , Inflammation/genetics , Inflammation/metabolism , Interleukin-10/metabolism , Lymphocyte Depletion , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Myelin-Oligodendrocyte Glycoprotein/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Signal Transduction , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocytes, Regulatory/metabolism
12.
J Exp Med ; 212(2): 217-33, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25646266

ABSTRACT

The co-stimulators ICOS (inducible T cell co-stimulator) and CD28 are both important for T follicular helper (TFH) cells, yet their individual contributions are unclear. Here, we show that each molecule plays an exclusive role at different stages of TFH cell development. While CD28 regulated early expression of the master transcription factor Bcl-6, ICOS co-stimulation was essential to maintain the phenotype by regulating the novel TFH transcription factor Klf2 via Foxo1. Klf2 directly binds to Cxcr5, Ccr7, Psgl-1, and S1pr1, and low levels of Klf2 were essential to maintain this typical TFH homing receptor pattern. Blocking ICOS resulted in relocation of fully developed TFH cells back to the T cell zone and reversion of their phenotype to non-TFH effector cells, which ultimately resulted in breakdown of the germinal center response. Our study describes for the first time the exclusive role of ICOS and its downstream signaling in the maintenance of TFH cells by controlling their anatomical localization in the B cell follicle.


Subject(s)
Gene Expression Regulation , Inducible T-Cell Co-Stimulator Protein/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Phenotype , T-Lymphocyte Subsets , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , CD28 Antigens/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Down-Regulation , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression , Germinal Center/immunology , Germinal Center/metabolism , Humans , Inducible T-Cell Co-Stimulator Ligand/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Palatine Tonsil/immunology , Palatine Tonsil/metabolism , Protein Binding , Receptors, CCR7/metabolism , Receptors, CXCR5/metabolism , Signal Transduction , T-Lymphocytes, Helper-Inducer/cytology
13.
Eur J Immunol ; 45(4): 975-87, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25639669

ABSTRACT

It is believed that memory CD8(+) T cells are maintained in secondary lymphoid tissues, peripheral tissues, and BM by homeostatic proliferation. Their survival has been shown to be dependent on IL-7, but it is unclear where they acquire it. Here we show that in murine BM, memory CD8(+) T cells individually colocalize with IL-7(+) reticular stromal cells. The T cells are resting in terms of global transcription and do not express markers of activation, for example, 4-1BB (CD137), IL-2, or IFN-γ, despite the expression of CD69 on about 30% of the cells. Ninety-five percent of the memory CD8(+) T cells in BM are in G0 phase of cell cycle and do not express Ki-67. Less than 1% is in S/M/G2 of cell cycle, according to propidium iodide staining. While previous publications have estimated the extent of proliferation of CD8(+) memory T cells on the basis of BrdU incorporation, we show here that BrdU itself induces proliferation of CD8(+) memory T cells. Taken together, the present results suggest that CD8(+) memory T cells are maintained as resting cells in the BM in dedicated niches with their survival conditional on IL-7 receptor signaling.


Subject(s)
Bone Marrow Cells/cytology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Resting Phase, Cell Cycle/immunology , Stromal Cells/immunology , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Bone Marrow Cells/immunology , Cell Proliferation , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Interleukin-7/immunology , Ki-67 Antigen/biosynthesis , Lectins, C-Type/biosynthesis , Mice , Mice, Inbred C57BL , Transcription, Genetic , Tumor Necrosis Factor Receptor Superfamily, Member 9/biosynthesis
14.
Blood ; 125(11): 1739-48, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25573986

ABSTRACT

Specific serum antibodies mediating humoral immunity and autoimmunity are provided by mature plasma cells (PC) residing in the bone marrow (BM), yet their dynamics and composition are largely unclear. We here characterize distinct subsets of human PC differing by CD19 expression. Unlike CD19(+) PC, CD19(-) PC were restricted to BM, expressed predominantly IgG, and they carried a prosurvival, distinctly mature phenotype, that is, HLA-DR(low)Ki-67(-)CD95(low)CD28(+)CD56(+/-), with increased BCL2 and they resisted their mobilization from the BM after systemic vaccination. Fewer mutations within immunoglobulin VH rearrangements of CD19(-) BMPC may indicate their differentiation in early life. Their resistance to in vivo B-cell depletion, that is, their independency from supply with new plasmablasts, is consistent with long-term stability of this PC subset in the BM. Moreover, CD19(-) PC were detectable in chronically inflamed tissues and secreted autoantibodies. We propose a multilayer model of PC memory in which CD19(+) and CD19(-) PC represent dynamic and static components, respectively, permitting both adaptation and stability of humoral immune protection.


Subject(s)
Antigens, CD19/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Immunoglobulin G/metabolism , Plasma Cells/immunology , Antilymphocyte Serum/administration & dosage , Bone Marrow Cells/classification , Boronic Acids/administration & dosage , Bortezomib , Cell Differentiation , Cell Survival , Diphtheria-Tetanus Vaccine/administration & dosage , Humans , Immunity, Humoral , Immunologic Memory , Inflammation/immunology , Inflammation/pathology , Lymphocyte Depletion , Models, Immunological , Mutation , Phenotype , Plasma Cells/classification , Plasma Cells/cytology , Pyrazines/administration & dosage , V(D)J Recombination
15.
Eur J Immunol ; 45(4): 1192-205, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25486906

ABSTRACT

Repeatedly activated T helper 1 (Th1) cells present during chronic inflammation can efficiently adapt to the inflammatory milieu, for example, by expressing the transcription factor Twist1, which limits the immunopathology caused by Th1 cells. Here, we show that in repeatedly activated murine Th1 cells, Twist1 and T-bet induce expression of microRNA-148a (miR-148a). miR-148a regulates expression of the proapoptotic gene Bim, resulting in a decreased Bim/Bcl2 ratio. Inhibition of miR-148a by antagomirs in repeatedly activated Th1 cells increases the expression of Bim, leading to enhanced apoptosis. Knockdown of Bim expression by siRNA in miR-148a antagomir-treated cells restores viability of the Th1 cells, demonstrating that miR-148a controls survival by regulating Bim expression. Thus, Twist1 and T-bet not only control the differentiation and function of Th1 cells, but also their persistence in chronic inflammation.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Gene Expression Regulation , Membrane Proteins/genetics , MicroRNAs/physiology , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/genetics , T-Box Domain Proteins/physiology , Th1 Cells/immunology , Twist-Related Protein 1/metabolism , Animals , Arthritis, Rheumatoid/immunology , Bcl-2-Like Protein 11 , Cell Survival/immunology , Cells, Cultured , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering , T-Box Domain Proteins/genetics , Twist-Related Protein 1/genetics
16.
Proc Natl Acad Sci U S A ; 111(25): 9229-34, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24927527

ABSTRACT

In the bone marrow, a population of memory T cells has been described that promotes efficient secondary immune responses and has been considered to be preactivated, owing to its expression of CD69 and CD25. Here we show that human bone marrow professional memory T cells are not activated but are resting in terms of proliferation, transcription, and mobility. They are in the G0 phase of the cell cycle, and their transcriptome is that of resting T cells. The repertoire of CD4(+) bone marrow memory T cells compared with CD4(+) memory T cells from the blood is significantly enriched for T cells specific for cytomegalovirus-pp65 (immunodominant protein), tetanus toxoid, measles, mumps, and rubella. It is not enriched for vaccinia virus and Candida albicans-MP65 (immunodominant protein), typical pathogens of skin and/or mucosa. CD4(+) memory T cells specific for measles are maintained nearly exclusively in the bone marrow. Thus, CD4(+) memory T cells from the bone marrow provide long-term memory for systemic pathogens.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Bone Marrow Cells/immunology , CD4-Positive T-Lymphocytes/immunology , Immunologic Memory/physiology , Interleukin-2 Receptor alpha Subunit/immunology , Lectins, C-Type/immunology , Resting Phase, Cell Cycle/immunology , Adult , Bone Marrow Cells/cytology , CD4-Positive T-Lymphocytes/cytology , Female , Humans , Male , Middle Aged
17.
Immunol Lett ; 160(2): 109-12, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24852107

ABSTRACT

B lymphocyte development in the mouse begins with the generation of long-term reconstituting, pluripotent hematopoietic stem cells, over multipotent myeloid/lymphoid progenitors and common lymphoid progenitors to B-lineage committed pro/pre B and pre B cells, which first express pre B cell receptors and then immunoglobulins, B cell receptors, to generate the repertoires of peripheral B cells. This development is influenced and guided by cells of non-hematopoietic and hematopoietic origins. We review here some of the recent developments, and our contributions in this fascinating field of developmental immunology.

19.
Eur J Immunol ; 44(6): 1615-21, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24643722

ABSTRACT

B-cell-derived interleukin-10 (IL-10) is known to act in a paracrine fashion to suppress inflammation. Here, we show that IL-10 also acts in an autocrine manner to regulate the differentiation of activated human B cells. We report that IL-10 expression is not restricted to a dedicated B-cell subset, but is induced transiently in peripheral human naïve, memory, and CD5(+) B cells alike upon activation. Global transcriptome comparison of activated human B cells, secreting IL-10 or not, identified 138 differentially regulated genes, most of which were associated with differentiation into antibody-secreting cells and reflecting autocrine IL-10 signaling. We monitored the differentiation of IL-10-secreting B cells and determined the effect of IL-10-blocking antibodies against its autocrine and paracrine signaling. IL-10 signaling promoted the differentiation of activated IL-10-secreting B cells into IgM- or IgG-secreting cells, but was dispensable for IgA secretion. Our data imply that B-cell-derived IL-10 not only suppresses immune reactions via paracrine mechanisms, but can also contribute to the differentiation of IL-10-secreting B cells into IgM- and IgG-secreting plasmablasts through both autocrine and paracrine signaling.


Subject(s)
Autocrine Communication/immunology , Cell Differentiation/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Interleukin-10/immunology , Paracrine Communication/immunology , Plasma Cells/immunology , Signal Transduction/physiology , Female , Humans , Immunoglobulin G/metabolism , Immunoglobulin M/metabolism , Immunologic Memory/physiology , Interleukin-10/metabolism , Male , Plasma Cells/cytology , Plasma Cells/metabolism
20.
Nature ; 507(7492): 366-370, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24572363

ABSTRACT

B lymphocytes have critical roles as positive and negative regulators of immunity. Their inhibitory function has been associated primarily with interleukin 10 (IL-10) because B-cell-derived IL-10 can protect against autoimmune disease and increase susceptibility to pathogens. Here we identify IL-35-producing B cells as key players in the negative regulation of immunity. Mice in which only B cells did not express IL-35 lost their ability to recover from the T-cell-mediated demyelinating autoimmune disease experimental autoimmune encephalomyelitis (EAE). In contrast, these mice displayed a markedly improved resistance to infection with the intracellular bacterial pathogen Salmonella enterica serovar Typhimurium as shown by their superior containment of the bacterial growth and their prolonged survival after primary infection, and upon secondary challenge, compared to control mice. The increased immunity found in mice lacking IL-35 production by B cells was associated with a higher activation of macrophages and inflammatory T cells, as well as an increased function of B cells as antigen-presenting cells (APCs). During Salmonella infection, IL-35- and IL-10-producing B cells corresponded to two largely distinct sets of surface-IgM(+)CD138(hi)TACI(+)CXCR4(+)CD1d(int)Tim1(int) plasma cells expressing the transcription factor Blimp1 (also known as Prdm1). During EAE, CD138(+) plasma cells were also the main source of B-cell-derived IL-35 and IL-10. Collectively, our data show the importance of IL-35-producing B cells in regulation of immunity and highlight IL-35 production by B cells as a potential therapeutic target for autoimmune and infectious diseases. This study reveals the central role of activated B cells, particularly plasma cells, and their production of cytokines in the regulation of immune responses in health and disease.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunity/immunology , Interleukins/metabolism , Salmonella Infections/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD40 Antigens/immunology , Female , Humans , Interleukin-10/metabolism , Interleukins/immunology , Lymphocyte Activation , Macrophages/cytology , Macrophages/immunology , Male , Mice , Plasma Cells/immunology , Plasma Cells/metabolism , Salmonella Infections/microbiology , T-Lymphocytes/immunology , Toll-Like Receptor 4/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...