Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Front Med (Lausanne) ; 10: 1221087, 2023.
Article in English | MEDLINE | ID: mdl-37663655

ABSTRACT

Objective: Chronic back pain (CBP) constitutes one of the most common complaints in primary care and a leading cause of disability worldwide. CBP may be of mechanical or inflammatory character and may lead to functional impairment and reduced quality of life. In this study, we aimed to assess and compare burden of disease, functional capacity, quality of life and depressive symptoms in axial spondyloarthritis (axSpA) patients with orthopedic chronic back pain patients (OBP). We further aimed to identify factors associated with quality of life. Methods: Cross-sectional survey of a cohort of 300 CBP patients including 150 patients from a University Hospital Orthopedic Back Pain Outpatient Clinic with OBP and 150 patients with confirmed axSpA from a University Hospital Rheumatology Outpatient Clinic. Questionnaire-based assessment of pain character (Inflammatory Back Pain, MAIL-Scale), functional status (FFbH, BASFI), quality of life (WHOQOL-Bref) and depressive symptoms (Phq9) and retrospective medical chart analysis. Results: Both, OBP and axSpA patients reported on average intermediate pain levels of mostly mixed pain character. Both groups demonstrated a reduced health-related quality of life and the presence of depressive symptoms. However, axSpA patients reported a significantly better subjective quality of life, more satisfaction with their health status and better functional capacity compared to OBP patients (all p < 0.001). In a multivariate regression model, depressive symptoms, mechanical back pain, pain level and age were negative predictors of subjective quality of life, whereas functional capacity was a positive predictor. Conclusion: Chronic back pain was associated with a high morbidity and reduced quality of life regardless of pain character. We identified multiple factors associated with reduced quality of life. Awareness and addressing of these factors may help to overcome unmet needs and improve quality of life for these patients.

3.
Front Immunol ; 13: 933191, 2022.
Article in English | MEDLINE | ID: mdl-36505422

ABSTRACT

Vitamin D (VD) deficiency is a highly prevalent worldwide phenomenon and is extensively discussed as a risk factor for the development of systemic lupus erythematosus (SLE) and other immune-mediated diseases. In addition, it is now appreciated that VD possesses multiple immunomodulatory effects. This study aims to explore the impact of dietary VD intake on lupus manifestation and pathology in lupus-prone NZB/W F1 mice and identify the underlying immunological mechanisms modulated by VD. Here, we show that low VD intake accelerates lupus progression, reflected in reduced overall survival and an earlier onset of proteinuria, as well higher concentrations of anti-double-stranded DNA autoantibodies. This unfavorable effect gained statistical significance with additional low maternal VD intake during the prenatal period. Among examined immunological effects, we found that low VD intake consistently hampered the adoption of a regulatory phenotype in lymphocytes, significantly reducing both IL-10-expressing and regulatory CD4+ T cells. This goes along with a mildly decreased frequency of IL-10-expressing B cells. We did not observe consistent effects on the phenotype and function of innate immune cells, including cytokine production, costimulatory molecule expression, and phagocytic capacity. Hence, our study reveals that low VD intake promotes lupus pathology, likely via the deviation of adaptive immunity, and suggests that the correction of VD deficiency might not only exert beneficial functions by preventing osteoporosis but also serve as an important module in prophylaxis and as an add-on in the treatment of lupus and possibly other immune-mediated diseases. Further research is required to determine the most appropriate dosage, as too-high VD serum levels may also induce adverse effects, possibly also on lupus pathology.


Subject(s)
Vitamin D Deficiency , Vitamin D , Animals , Mice , Female , Pregnancy , Interleukin-10 , Mice, Inbred NZB , Vitamins , Diet
4.
EMBO Mol Med ; 14(1): e14182, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34842342

ABSTRACT

Fc-gamma receptor (FcγR) activation by soluble IgG immune complexes (sICs) represents a major mechanism of inflammation in certain autoimmune diseases such as systemic lupus erythematosus (SLE). A robust and scalable test system allowing for the detection and quantification of sIC bioactivity is missing. We developed a comprehensive reporter cell panel detecting activation of FcγRs. The reporter cell lines were integrated into an assay that enables the quantification of sIC reactivity via ELISA or a faster detection using flow cytometry. This identified FcγRIIA(H) and FcγRIIIA as the most sIC-sensitive FcγRs in our test system. Reaching a detection limit in the very low nanomolar range, the assay proved also to be sensitive to sIC stoichiometry and size reproducing for the first time a complete Heidelberger-Kendall curve in terms of immune receptor activation. Analyzing sera from SLE patients and mouse models of lupus and arthritis proved that sIC-dependent FcγR activation has predictive capabilities regarding severity of SLE disease. The assay provides a sensitive and scalable tool to evaluate the size, amount, and bioactivity of sICs in all settings.


Subject(s)
Lupus Erythematosus, Systemic , Receptors, IgG , Animals , Antigen-Antibody Complex/metabolism , Flow Cytometry , Humans , Inflammation , Mice , Receptors, IgG/metabolism
5.
Front Immunol ; 12: 765741, 2021.
Article in English | MEDLINE | ID: mdl-34925335

ABSTRACT

A link between high sodium chloride (salt) intake and the development of autoimmune diseases was previously reported. These earlier studies demonstrated exacerbation of experimental autoimmune encephalomyelitis and colitis by excess salt intake associated with Th17- and macrophage-mediated mechanisms. Little is known about the impact of dietary salt intake on experimental arthritides. Here, we investigated if salt restriction can exert beneficial effects on collagen-induced arthritis (CIA) and K/BxN serum transfer-induced arthritis (STIA). CIA depends on both adaptive and innate immunity, while STIA predominantly mimics the innate immune cell-driven effector phase of arthritis. In both models, low salt (LS) diet significantly decreased arthritis severity compared to regular salt (RS) and high salt (HS) diet. We did not observe an aggravation of arthritis with HS diet compared to RS diet. Remarkably, in STIA, LS diet was as effective as IL-1 receptor blocking treatment. Complement-fixing anti-CII IgG2a antibodies are associated with inflammatory cell infiltration and cartilage destruction. LS diet reduced anti-CII IgG2a levels in CIA and decreased the anti-CII IgG2a/IgG1 ratios pointing toward a more Th2-like response. Significantly less inflammatory joint infiltrates and cartilage breakdown associated with reduced protein concentrations of IL-1 beta (CIA and STIA), IL-17 (CIA), and the monocyte chemoattractant protein-1 (MCP-1) (CIA) were detected in mice receiving LS diet compared to HS diet. However, we did not find a reduced IL-17A expression in CD4+ T cells upon salt restriction in CIA. Analysis of mRNA transcripts and immunoblots revealed a link between LS diet and inhibition of the p38 MAPK (mitogen-activated protein kinase)/NFAT5 (nuclear factor of activated T-cells 5) signaling axis in STIA. Further experiments indicated a decreased leukodiapedesis under LS conditions. In conclusion, dietary salt restriction ameliorates CIA and STIA, indicating a beneficial role of LS diet during both the immunization and effector phase of immune-mediated arthritides by predominantly modulating the humoral immunity and the activation status of myeloid lineage cells. Hence, salt restriction might represent a supportive dietary intervention not only to reduce cardiovascular risk, but also to improve human inflammatory joint diseases like rheumatoid arthritis.


Subject(s)
Arthritis, Experimental , Diet, Sodium-Restricted , Adaptive Immunity , Animals , Arthritis, Experimental/blood , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , B-Lymphocytes/immunology , Cytokines/genetics , Cytokines/immunology , E-Selectin/immunology , Endothelial Cells/immunology , Foot Joints/immunology , Foot Joints/pathology , Immunity, Innate , Immunoglobulin G/blood , Mice, Inbred C57BL , Mice, Inbred DBA , Monocytes/immunology , Myeloid Progenitor Cells/immunology , Receptors, Interleukin-1/immunology
6.
Front Immunol ; 12: 696810, 2021.
Article in English | MEDLINE | ID: mdl-34335609

ABSTRACT

Changed dietary habits in Western countries such as reduced fiber intake represent an important lifestyle factor contributing to the increase in inflammatory immune-mediated diseases. The mode of action of beneficial fiber effects is not fully elucidated, but short-chain fatty acids (SCFA) and gut microbiota have been implicated. The aim of this study was to explore the impact of dietary fiber on lupus pathology and to understand underlying mechanisms. Here, we show that in lupus-prone NZB/WF1 mice low fiber intake deteriorates disease progression reflected in accelerated mortality, autoantibody production and immune dysregulation. In contrast to our original assumption, microbiota suppression by antibiotics or direct SCFA feeding did not influence the course of lupus-like disease. Mechanistically, our data rather indicate that in low fiber-fed mice, an increase in white adipose tissue mass, fat-inflammation and a disrupted intestinal homeostasis go along with systemic, low-grade inflammation driving autoimmunity. The links between obesity, intestinal leakage and low-grade inflammation were confirmed in human samples, while adaptive immune activation predominantly correlated with lupus activity. We further propose that an accelerated gastro-intestinal passage along with energy dilution underlies fiber-mediated weight regulation. Thus, our data highlight the often-overlooked effects of dietary fiber on energy homeostasis and obesity prevention. Further, they provide insight into how intricately the pathologies of inflammatory immune-mediated conditions, such as obesity and autoimmunity, might be interlinked, possibly sharing common pathways.


Subject(s)
Dietary Fiber/deficiency , Lupus Erythematosus, Systemic/etiology , Obesity/etiology , Adaptive Immunity , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adiposity , Adolescent , Adult , Aged , Aged, 80 and over , Animal Feed , Animals , Autoantibodies/blood , Autoimmunity , Case-Control Studies , Dietary Fiber/administration & dosage , Disease Models, Animal , Disease Progression , Energy Metabolism , Female , Humans , Inflammation Mediators/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Male , Mice, Inbred NZB , Middle Aged , Nutritive Value , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Permeability , Young Adult
7.
Front Immunol ; 12: 673912, 2021.
Article in English | MEDLINE | ID: mdl-34108971

ABSTRACT

Objectives: ANCA-associated vasculitides (AAV) affect small- and medium-sized blood vessels. In active disease, vessel wall infiltrates are mainly composed of monocytes and macrophages. Immune checkpoint molecules are crucial for the maintenance of self-tolerance and the prevention of autoimmune diseases. After checkpoint inhibitor therapy, the development of autoimmune vasculitis has been observed. However, defects of immune checkpoint molecules in AAV patients have not been identified yet. Methods: Monocytes and monocyte-derived macrophages from AAV patients and healthy age-matched controls were tested for surface expression of immunoinhibitory checkpoint programmed cell death ligand-1 (PD-L1). Using in vitro co-culture approaches, the effect of monocyte PD-L1 expression on CD4+ T cell activation and proliferation was tested. Results: Monocytes from AAV patients displayed lower PD-L1 expression and a defective PD-L1 presentation upon activation, an effect that was correlated with disease activity. Lower PD-L1 expression was due to increased lysosomal degradation of PD-L1 in AAV monocytes. We identified a reduced expression of CMTM6, a protein protecting PD-L1 from lysosomal breakdown, as the underlying molecular defect. PD-L1low AAV monocytes showed increased stimulatory capacity and induced T cell activation and proliferation. Inhibiting lysosomal function corrected this phenotype by increasing PD-L1, thus normalizing the pro-stimulatory behavior of AAV monocytes. Conclusions: This study identifies a defect of the immunoinhibitory checkpoint PD-L1 in monocytes from patients with AAV. Low expression of CMTM6 results in enhanced lysosomal degradation of PD-L1, thus providing insufficient negative signaling to T cells. Correcting this defect by targeting lysosomal function may represent a novel strategy to treat AAV.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/immunology , B7-H1 Antigen/immunology , Lymphocyte Activation/immunology , MARVEL Domain-Containing Proteins/metabolism , Monocytes/immunology , Myelin Proteins/metabolism , Aged , Antigen Presentation/immunology , CD4-Positive T-Lymphocytes/immunology , Female , Humans , Male , Middle Aged , Monocytes/metabolism
8.
Int J Mol Sci ; 22(3)2021 Jan 29.
Article in English | MEDLINE | ID: mdl-33572870

ABSTRACT

As a key anti-inflammatory cytokine, IL-10 is crucial in preventing inflammatory and autoimmune diseases. However, in human and murine lupus, its role remains controversial. Our aim was to understand regulation and immunologic effects of IL-10 on different immune functions in the setting of lupus. This was explored in lupus-prone NZB/W F1 mice in vitro and vivo to understand IL-10 effects on individual immune cells as well as in the complex in vivo setting. We found pleiotropic IL-10 expression that largely increased with progressing lupus, while IL-10 receptor (IL-10R) levels remained relatively stable. In vitro experiments revealed pro- and anti-inflammatory IL-10 effects. Particularly, IL-10 decreased pro-inflammatory cytokines and slowed B cell proliferation, thereby triggering plasma cell differentiation. The frequent co-expression of ICOS, IL-21 and cMAF suggests that IL-10-producing CD4 T cells are important B cell helpers in this context. In vitro and in vivo effects of IL-10 were not fully concordant. In vivo IL-10R blockade slightly accelerated clinical lupus manifestations and immune dysregulation. Altogether, our side-by-side in vitro and in vivo comparison of the influence of IL-10 on different aspects of immunity shows that IL-10 has dual effects. Our results further reveal that the overall outcome may depend on the interplay of different factors such as target cell, inflammatory and stimulatory microenvironment, disease model and state. A comprehensive understanding of such influences is important to exploit IL-10 as a therapeutic target.


Subject(s)
Interleukin-10/immunology , Lupus Erythematosus, Systemic/immunology , Adaptive Immunity , Animals , Autoimmunity , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Disease Models, Animal , Disease Progression , Female , Gene Expression , Humans , Immunity, Innate , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-10/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Mice, Inbred NZB , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
9.
Int J Mol Sci ; 20(13)2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31266174

ABSTRACT

It is incompletely understood how self-antigens become targets of humoral immunity in antibody-mediated autoimmune diseases. In this context, alarmins are discussed as an important level of regulation. Alarmins are recognized by various receptors, such as receptor for advanced glycation end products (RAGE). As RAGE is upregulated under inflammatory conditions, strongly binds nucleic acids and mediates pro-inflammatory responses upon alarmin recognition, our aim was to examine its contribution to immune complex-mediated autoimmune diseases. This question was addressed employing RAGE-/- animals in murine models of pristane-induced lupus, collagen-induced, and serum-transfer arthritis. Autoantibodies were assessed by enzyme-linked immunosorbent assay, renal disease by quantification of proteinuria and histology, arthritis by scoring joint inflammation. The associated immune status was determined by flow cytometry. In both disease entities, we detected tendentiously decreased autoantibody levels in RAGE-/- mice, however no differences in clinical outcome. In accordance with autoantibody levels, a subgroup of the RAGE-/- animals showed a decrease in plasma cells, and germinal center B cells and an increase in follicular B cells. Based on our results, we suggest that RAGE deficiency alone does not significantly affect antibody-mediated autoimmunity. RAGE may rather exert its effects along with other receptors linking environmental factors to auto-reactive immune responses.


Subject(s)
Arthritis, Experimental/immunology , Autoantibodies/metabolism , Lupus Nephritis/immunology , Receptor for Advanced Glycation End Products/deficiency , Animals , Arthritis, Experimental/genetics , Autoantibodies/blood , B-Lymphocytes/immunology , Collagen/adverse effects , Disease Models, Animal , Germinal Center/immunology , Lupus Nephritis/genetics , Mice , Receptor for Advanced Glycation End Products/immunology , Terpenes/adverse effects
10.
J Immunol Methods ; 474: 112628, 2019 11.
Article in English | MEDLINE | ID: mdl-31254500

ABSTRACT

Plasma cells (PCs) secrete antibodies and play an essential role in protective immunity, but also in pathogenesis of antibody-mediated diseases. Physiologically, PCs mainly reside within bone marrow and spleen. In autoimmune diseases such as systemic lupus erythematosus (SLE) autoantibody-producing PCs can also be found at sites of inflammation, e.g. in nephritic kidneys. Therefore, efficient methods are required to reliably analyze and compare PCs at different sites. Flow cytometry and ELISpot analyses are frequently employed for PC characterization and require the preparation of single cell suspensions. To that end, enzymatic digestion is commonly used to isolate immune cells from solid organs like kidneys, occasionally also from lymphoid organs. In this study we show that enzymatic digestion using collagenase may lead to a loss of certain surface markers, e.g. the PC markers CD138 and CD267 (TACI). Therefore, we established an optimized protocol for preparing renal single cells by merely applying mechanical tissue disruption. Omitting enzymatic digestion, this method enables a reliable characterization of viable renal PCs by flow cytometry and cell sorting. We further show that mechanic cell preparation is favorable for lymphocytic immune cell enrichment, while enzymatic disruption improves the yield of digitating or stroma cell populations.


Subject(s)
Cell Separation/methods , Dissection , Flow Cytometry , Kidney/immunology , Plasma Cells/immunology , Animals , Biomarkers/metabolism , Cell Survival , Collagenases/metabolism , Female , Kidney/cytology , Kidney/metabolism , Mice, Inbred MRL lpr , Mice, Inbred NZB , Plasma Cells/metabolism , Time Factors , Workflow
11.
J Autoimmun ; 73: 120-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27427404

ABSTRACT

Environmental factors contribute to development of autoimmune diseases. For instance, human autoimmune arthritis can associate with intestinal inflammation, cigarette smoking, periodontal disease, and various infections. The cellular and, molecular pathways whereby such remote challenges might precipitate arthritis or flares remain unclear. Here, we used a transfer model of self-reactive arthritis-inducing CD4(+) cells from KRNtg mice that, upon transfer, induce a very mild form of autoinflammatory arthritis in recipient animals. This model enabled us to identify external factors that greatly aggravated disease. We show that several distinct challenges precipitated full-blown arthritis, including intestinal inflammation through DSS-induced colitis, and bronchial stress through Influenza infection. Both triggers induced strong IL-17 expression primarily in self-reactive CD4(+) cells in lymph nodes draining the site of inflammation. Moreover, treatment of mice with IL-1ß greatly exacerbated arthritis, while transfer of KRNtg CD4(+) cells lacking IL-1R significantly reduced disease and IL-17 expression. Thus, IL-1ß enhances the autoaggressive potential of self-reactive CD4(+) cells, through increased Th17 differentiation, and this influences inflammatory events in the joints. We propose that diverse challenges that cause remote inflammation (lung infection or colitis, etc.) result in IL-1ß-driven Th17 differentiation, and this precipitates arthritis in genetically susceptible individuals. Thus the etiology of autoimmune inflammatory arthritis likely relates to diverse triggers that converge to a common pathway involving IL-1ß production and Th17 cell distribution.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Interleukin-1beta/metabolism , Spondylarthritis/immunology , Th17 Cells/immunology , Adoptive Transfer , Animals , Arthritis, Rheumatoid/genetics , Colitis/chemically induced , Colitis/immunology , Dextran Sulfate/toxicity , Genetic Predisposition to Disease , Influenza A virus/immunology , Interleukin-17/metabolism , Joints/immunology , Klebsiella pneumoniae/immunology , Lung Diseases/immunology , Lung Diseases/virology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Th17 Cells/metabolism
12.
Leuk Lymphoma ; 57(9): 2150-60, 2016 09.
Article in English | MEDLINE | ID: mdl-26757600

ABSTRACT

The immune cell composition of the follicular lymphoma (FL) tumor microenvironment is increasingly recognized as an important determinant for clinical outcome. Here, we explored frequency and distribution of dendritic cell (DC) subtypes in relation to regulatory T cells (Treg) by immunohistochemistry in lymph node biopsies from patients with de novo FL. We found that neoplastic follicles contained lower DC and higher Treg frequencies than hyperplastic follicles in control lymph nodes. Treg numbers particularly correlated with the subset of conventional CD11c(+ )DCs. Additionally, both a high intra- to interfollicular ratio of CD11c(+ )DCs and increased intrafollicular Treg frequencies were associated with decreased overall survival. This suggests that functional interactions between these cells may be relevant for FL progression/recurrence. The presence of CD11c(+ )DCs in the tumor microenvironment may assist tumor infiltration by Tregs, thus contributing to the suppression of an otherwise beneficial T-cell-dominated FL microenvironment.


Subject(s)
Dendritic Cells/immunology , Lymphoma, Follicular/immunology , Lymphoma, Follicular/pathology , Tumor Microenvironment/immunology , Adult , Aged , Aged, 80 and over , Biopsy , Dendritic Cells/metabolism , Dendritic Cells/pathology , Disease Progression , Female , Gene Expression , HLA-DR Antigens/genetics , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymph Nodes/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Lymphoma, Follicular/mortality , Lymphoma, Follicular/therapy , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
13.
Arthritis Rheumatol ; 68(4): 1026-38, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26501485

ABSTRACT

OBJECTIVE: Antibody-mediated autoimmunity involves cognate interactions between self-reactive T cells and B cells during germinal center (GC) reactions. The aim of this study was to determine the role of essential follicular helper T (Tfh) cell molecules (CXCR5, signaling lymphocytic activation molecule-associated protein) on autoreactive CD4+ cells and the role of certain environmental influences that may determine GC-driven autoantibody production and arthritis development. METHODS: We transferred self-reactive CD4+ cells from KRN-Tg mice into recipient mice, which induced autoantibodies and autoinflammatory arthritis. This model allowed manipulation of environmental effects, such as inflammation, and use of transferred cells that were genetically deficient in important Tfh cell-associated molecules. RESULTS: A deficiency of signaling lymphocytic activation molecule-associated protein (SAP) in CD4+ cells from KRN-Tg mice completely protected against arthritis, indicating that stable T cell-B cell interactions are required for GC formation, autoantibody production, and arthritis induction. In contrast, a CXCR5 deficiency in CD4+ cells from KRN-Tg mice still induced disease when these cells were transferred into wild-type mice, suggesting that T cell help for B cells could rely on other migration mechanisms. However, various manipulations influenced this system, including elimination of bystander effects through use of CD28(-/-) recipient mice (reduced disease) or use of inflammation-inducing Freund's complete adjuvant (progression to arthritis). We also examined the capacity of preexisting GCs with a nonautoimmune specificity to co-opt autoimmune T cells and observed no evidence for any influence. CONCLUSION: In addition to the quality and quantity of cognate CD4+ cell help, external factors such as inflammation and noncognate CD4+ cell bystander activation trigger autoimmunity by shaping events within autoimmune GC responses. SAP is an essential molecule for autoimmune antibody production, whereas the importance of CXCR5 varies depending on the circumstances.


Subject(s)
Arthritis, Rheumatoid/immunology , Autoantibodies/biosynthesis , Environment , Intracellular Signaling Peptides and Proteins/immunology , Receptors, CXCR5/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Arthritis, Psoriatic/immunology , Arthritis, Rheumatoid/genetics , Autoantibodies/immunology , Autoimmune Diseases/immunology , Disease Models, Animal , Disease Progression , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Germinal Center/cytology , Glucose-6-Phosphate Isomerase/immunology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Receptors, CXCR5/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signaling Lymphocytic Activation Molecule Associated Protein
14.
Nat Commun ; 6: 7320, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26102221

ABSTRACT

Asthma is prevalent in Western countries, and recent explanations have evoked the actions of the gut microbiota. Here we show that feeding mice a high-fibre diet yields a distinctive gut microbiota, which increases the levels of the short-chain fatty acid, acetate. High-fibre or acetate-feeding led to marked suppression of allergic airways disease (AAD, a model for human asthma), by enhancing T-regulatory cell numbers and function. Acetate increases acetylation at the Foxp3 promoter, likely through HDAC9 inhibition. Epigenetic effects of fibre/acetate in adult mice led us to examine the influence of maternal intake of fibre/acetate. High-fibre/acetate feeding of pregnant mice imparts on their adult offspring an inability to develop robust AAD. High fibre/acetate suppresses expression of certain genes in the mouse fetal lung linked to both human asthma and mouse AAD. Thus, diet acting on the gut microbiota profoundly influences airway responses, and may represent an approach to prevent asthma, including during pregnancy.


Subject(s)
Acetates/metabolism , Asthma/metabolism , Diet , Dietary Fiber/metabolism , Gastrointestinal Microbiome , Prenatal Exposure Delayed Effects/metabolism , T-Lymphocytes, Regulatory/immunology , Acetates/pharmacology , Acetylation/drug effects , Animals , Asthma/immunology , Disease Models, Animal , Epigenesis, Genetic/drug effects , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/pharmacology , Female , Forkhead Transcription Factors/drug effects , Forkhead Transcription Factors/genetics , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Mice , Pregnancy , Prenatal Exposure Delayed Effects/immunology , Promoter Regions, Genetic , Repressor Proteins/drug effects , Repressor Proteins/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects
15.
Nat Commun ; 6: 6734, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25828455

ABSTRACT

Diet and the gut microbiota may underpin numerous human diseases. A major metabolic product of commensal bacteria are short-chain fatty acids (SCFAs) that derive from fermentation of dietary fibre. Here we show that diets deficient or low in fibre exacerbate colitis development, while very high intake of dietary fibre or the SCFA acetate protects against colitis. SCFAs binding to the 'metabolite-sensing' receptors GPR43 and GPR109A in non-haematopoietic cells mediate these protective effects. The inflammasome pathway has hitherto been reported as a principal pathway promoting gut epithelial integrity. SCFAs binding to GPR43 on colonic epithelial cells stimulates K(+) efflux and hyperpolarization, which lead to NLRP3 inflammasome activation. Dietary fibre also shapes gut bacterial ecology, resulting in bacterial species that are more effective for inflammasome activation. SCFAs and metabolite receptors thus explain health benefits of dietary fibre, and how metabolite signals feed through to a major pathway for gut homeostasis.


Subject(s)
Carrier Proteins/drug effects , Colitis/metabolism , Colon/drug effects , Dietary Fiber/pharmacology , Fatty Acids, Volatile/metabolism , Inflammasomes/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/metabolism , Acetates/metabolism , Animals , Butyrates/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Colon/metabolism , Fermentation , Gastrointestinal Microbiome , Homeostasis/drug effects , Inflammasomes/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, G-Protein-Coupled/genetics , Receptors, Nicotinic/genetics
16.
Methods Mol Biol ; 1291: 209-26, 2015.
Article in English | MEDLINE | ID: mdl-25836314

ABSTRACT

Blood-circulating CXCR5(+) CD4(+) T cells and T follicular helper (TFH) cells, which participate in germinal center (GC) reactions within secondary lymphoid organs, are specialized in providing help to B cells. This chapter describes ways to isolate TFH-like cells out of peripheral blood or tonsils and to quantify their B cell helper function. This comprises different co-culture approaches of TFH-like cells and B cells and the evaluation of their capacity to induce immunoglobulin secretion and plasma cell differentiation. In addition, B cell helper function of CD4 T cells can be estimated indirectly by quantifying the expression of B cell helper cytokines and co-stimulatory and TFH-associated molecules.


Subject(s)
Immunologic Techniques/methods , T-Lymphocytes, Helper-Inducer/cytology , B-Lymphocytes/cytology , Cell Separation , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunomagnetic Separation , Lymphocyte Activation/immunology , Palatine Tonsil/cytology , Phenotype , Real-Time Polymerase Chain Reaction
17.
J Immunol ; 193(10): 4845-58, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25339665

ABSTRACT

The dynamic interplay between regulatory T cells (T(regs)) and effector T cells (T(effs)) governs the balance between tolerance and effector immune responses. Perturbations of T(reg) frequency and function or imbalances in T(reg)/T(eff) levels are associated with the development of autoimmunity. The factors that mediate these changes remain poorly understood and were investigated in this study in murine autoimmune arthritis. T(regs) displayed a stable phenotype in arthritic mice and were fully functional in in vitro suppression assays. However, their expansion was delayed relative to T(effs) (T follicular helper cells and Th17 cells) during the early stages of autoimmune reactivity. This imbalance is likely to have led to insufficient T(reg) control of T(effs) and induced autoimmunity. Moreover, a counterregulatory and probably IL-7-driven increase in thymic T(reg) production and recruitment to inflamed tissues was too slow for disease prevention. Increased T(eff) over T(reg) expansion was further aggravated by inflammation and lymphopenia. Both these conditions contribute to autoimmune pathogenesis and were accompanied by decreases in the availability of IL-2 and increases in levels of IL-21. IL-2 neutralization or supplementation was used to show that T(reg) expansion mainly depended on this cytokine. IL-21R(-/-) cells were used to demonstrate that IL-21 promoted the maintenance of T(effs). Thus, at inflammatory sites in experimental arthritis, a deficit in IL-2 hampers T(reg) proliferation, whereas exaggerated IL-21 levels overwhelm T(reg) control by supporting T(eff) expansion. This identifies IL-2 and IL-21 as targets for manipulation in therapies for autoimmunity.


Subject(s)
Arthritis/immunology , Autoimmunity , Interleukin-2/immunology , Interleukins/immunology , Lymphopenia/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Arthritis/complications , Arthritis/genetics , Arthritis/pathology , Cell Proliferation , Gene Expression Regulation , Immune Tolerance , Inflammation/complications , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-2/genetics , Interleukins/genetics , Lymphopenia/complications , Lymphopenia/genetics , Lymphopenia/pathology , Mice , Mice, Transgenic , Receptors, Interleukin-21/genetics , Receptors, Interleukin-21/immunology , Signal Transduction , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Regulatory/pathology
18.
Immunity ; 39(4): 770-81, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24138884

ABSTRACT

Follicular B helper T (Tfh) cells support high affinity and long-term antibody responses. Here we found that within circulating CXCR5⁺ CD4⁺ T cells in humans and mice, the CCR7(lo)PD-1(hi) subset has a partial Tfh effector phenotype, whereas CCR7(hi)PD-1(lo) cells have a resting phenotype. The circulating CCR7(lo)PD-1(hi) subset was indicative of active Tfh differentiation in lymphoid organs and correlated with clinical indices in autoimmune diseases. Thus the CCR7(lo)PD-1(hi) subset provides a biomarker to monitor protective antibody responses during infection or vaccination and pathogenic antibody responses in autoimmune diseases. Differentiation of both CCR7(hi)PD-1(lo) and CCR7(lo)PD-1(hi) subsets required ICOS and BCL6, but not SAP, suggesting that circulating CXCR5⁺ helper T cells are primarily generated before germinal centers. Upon antigen reencounter, CCR7(lo)PD-1(hi) CXCR5⁺ precursors rapidly differentiate into mature Tfh cells to promote antibody responses. Therefore, circulating CCR7(lo)PD-1(hi) CXCR5⁺ CD4⁺ T cells are generated during active Tfh differentiation and represent a new mechanism of immunological early memory.


Subject(s)
Antibodies/immunology , Immunologic Memory , Programmed Cell Death 1 Receptor/immunology , Receptors, CXCR5/immunology , Receptors, CXCR/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigens/immunology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , B-Lymphocytes/virology , Cell Differentiation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Gene Expression , Germinal Center/immunology , Germinal Center/pathology , Germinal Center/virology , Humans , Immunity, Humoral , Immunophenotyping , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/immunology , Mice , Programmed Cell Death 1 Receptor/genetics , Proto-Oncogene Proteins c-bcl-6 , Receptors, CXCR/genetics , Receptors, CXCR5/genetics , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Helper-Inducer/virology
19.
J Immunol ; 191(8): 4112-20, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24048894

ABSTRACT

The induction of regulatory T cells (Tregs) to suppress aberrant inflammation and immunity has potential as a therapeutic strategy for asthma. Recently, we identified key immunoregulatory components of Streptococcus pneumoniae, type 3 polysaccharide and pneumolysoid (T+P), which suppress allergic airways disease (AAD) in mouse models of asthma. To elucidate the mechanisms of suppression, we have now performed a thorough examination of the role of Tregs. BALB/c mice were sensitized to OVA (day 0) i.p. and challenged intranasal (12-15 d later) to induce AAD. T+P was administered intratracheally at the time of sensitization in three doses (0, 12, and 24 h). T+P treatment induced an early (36 h-4 d) expansion of Tregs in the mediastinal lymph nodes, and later (12-16 d) increases in these cells in the lungs, compared with untreated allergic controls. Anti-CD25 treatment showed that Treg-priming events involving CD25, CCR7, IL-2, and TGF-ß were required for the suppression of AAD. During AAD, T+P-induced Tregs in the lungs displayed a highly suppressive phenotype and had an increased functional capacity. T+P also blocked the induction of IL-6 to prevent the Th17 response, attenuated the expression of the costimulatory molecule CD86 on myeloid dendritic cells (DCs), and reduced the number of DCs carrying OVA in the lung and mediastinal lymph nodes. Therefore, bacterial components (T+P) drive the differentiation of highly suppressive Tregs, which suppress the Th2 response, prevent the Th17 response and disable the DC response resulting in the effective suppression of AAD.


Subject(s)
Asthma/immunology , Polysaccharides, Bacterial/immunology , Streptococcus pneumoniae/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B7-2 Antigen/biosynthesis , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Inflammation/immunology , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-6/biosynthesis , Lung/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Polysaccharides, Bacterial/administration & dosage , Receptors, CCR7/metabolism , Th17 Cells/immunology , Th2 Cells/immunology , Transforming Growth Factor beta/metabolism
20.
J Immunol ; 186(10): 5556-68, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21471443

ABSTRACT

High expression of CXCR5 is one of the defining hallmarks of T follicular helper cells (T(FH)), a CD4 Th cell subset that promotes germinal center reactions and the selection and affinity maturation of B cells. CXCR5 is also expressed on 20-25% of peripheral blood human central memory CD4 T cells (T(CM)), although the definitive function of these cells is not fully understood. The constitutive expression of CXCR5 on T(FH) cells and a fraction of circulating T(CM) suggests that CXCR5(+) T(CM) may represent a specialized subset of memory-type T(FH) cells programmed for homing to follicles and providing B cell help. To verify this assumption, we analyzed this cell population and show its specialized function in supporting humoral immune responses. Compared with their CXCR5(-) T(CM) counterparts, CXCR5(+) T(CM) expressed high levels of the chemokine CXCL13 and efficiently induced plasma cell differentiation and Ig secretion. We found that the distinct B cell helper qualities of CXCR5(+) T(CM) were mainly due to high ICOS expression and pronounced responsiveness to ICOS ligand costimulation together with large IL-10 secretion. Furthermore, B cell helper attributes of CXCR5(+) T(CM) were almost exclusively acquired on cognate interaction with B cells, but not with dendritic cells. This implies that a preferential recruitment of circulating CXCR5(+) T(CM) to CXCL13-rich B cell follicles is required for the promotion of a quick and efficient protective secondary humoral immune response. Taken together, we propose that CXCR5(+) T(CM) represent a distinct memory cell subset specialized in supporting Ab-mediated immune responses.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Immunity, Humoral , Immunologic Memory , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , T-Lymphocyte Subsets/immunology , Antigens, Differentiation, T-Lymphocyte/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Dendritic Cells , Enzyme-Linked Immunosorbent Assay , Humans , Inducible T-Cell Co-Stimulator Protein , Interleukin-10/biosynthesis , Interleukin-10/immunology , Interleukin-10/metabolism , Lymphocyte Activation , Microarray Analysis , Palatine Tonsil/immunology , Polymerase Chain Reaction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...