Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Int J Mol Sci ; 24(18)2023 Sep 16.
Article in English | MEDLINE | ID: mdl-37762476

ABSTRACT

It is well established that the neonatal immune system is different from the adult immune system. A major task of the neonatal immune system is to bridge the achievement of tolerance towards harmless antigens and commensal bacteria while providing protection against pathogens. This is highly important because neonates are immunologically challenged directly after birth by a rigorous change from a semi-allogeneic sterile environment into a world rich with microbes. A so called disease tolerogenic state is typical for neonates and is anticipated to prevent immunopathological damage potentially at the cost of uncontrolled pathogen proliferation. As a consequence, neonates are more susceptible than adults to life-threatening infections. At the basis of a well-functioning immune response, both for adults and neonates, innate immune cells such as monocytes and monocyte-derived macrophages play an essential role. A well-responsive monocyte will alter its cellular metabolism to subsequently induce certain immune effector function, a process which is called immunometabolism. Immunometabolism has received extensive attention in the last decade; however, it has not been broadly studied in neonates. This review focuses on carbohydrate metabolism in monocytes and macrophages in neonates. We will exhibit pathways involving glycolysis, the tricarboxylic acid (TCA) cycle and oxidative phosphorylation and their role in shaping neonates' immune systems to a favorable tolerogenic state. More insight into these pathways will elucidate potential treatments targets in life-threatening conditions including neonatal sepsis or expose potential targets which can be used to induce tolerance in conditions where tolerance is harmfully impaired such as in autoimmune diseases.


Subject(s)
Autoimmune Diseases , Immune Tolerance , Monocytes , Adult , Humans , Infant, Newborn , Citric Acid Cycle , Macrophages
2.
Front Pediatr ; 11: 1123873, 2023.
Article in English | MEDLINE | ID: mdl-37456566

ABSTRACT

Objectives: CD4+ memory T cells facilitate long-termed adaptive immune responses while NK cells are predominately rapid effector cells with significant functions for both intestinal homeostasis and inflammation. We wanted to study both populations in health and pediatric inflammatory bowel disease (IBD) and correlate them with disease activity and medication. Methods: We performed flow cytometric analyses of peripheral blood CD4 + CD45RO+ memory T cells and CD3-CD16 + CD56+ NK cells in 30 patients with IBD and 31 age-matched controls and correlated percentages of subsets with disease activity (PUCAI/PCDAI) and medication. Results: We found a significant reduction of peripheral NK cells in overall IBD patients with both clinical remission and disease activity, which was even more pronounced in patients treated with azathioprine. Otherwise, circulating CD4+ memory T cell populations were significantly enhanced in active IBD compared to controls. Enhancement of memory T cells was particularly found in new onset disease and correlated with disease activity scores. Discussion: Our single center cohort confirms previous results showing enhanced memory T cell populations in pediatric IBD patients, which correlate with disease activity scores. CD4+ memory T cells are a relevant pathogenic leukocyte population for disease development and perpetuation in IBD. In addition, we found a decrease of NK cells in IBD patients, which was pronounced by use of azathioprine. Surveillance of both cellular populations could possibly serve as biomarker for therapy control in pediatric IBD.

3.
Sci Rep ; 13(1): 3762, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36882462

ABSTRACT

Tofactinib is a JAK inhibitor approved for ulcerative colitis in humans. Despite of its' proven effectiveness in humans, mechanistic data are scarce on the effectiveness of Tofactinib in experimental colitis in mice. We induced experimental colitis by transfer of CD4+CD25- isolated T cells into RAG2-/- (T and B cell deficient) mice and treated these mice with tofacitinib for 5-6 weeks either with a dosage of 10 or 40 mg/kg body weight immediately after CD4+ transfer or started treatment after first symptoms of disease for several weeks. While treatment with tofacitinib immediately after transfer resulted in an enhanced expansion of CD4+ T cells and did not prevent occurrence of colitis, treatment after start of symptoms of colitis ameliorated disease activity on a clinical basis and in histological analyses. Tofacitinib is effective in the treatment of murine experimental T cell transfer colitis, however does not prevent occurrence of disease.


Subject(s)
Colitis, Ulcerative , Colitis , Humans , Animals , Mice , Colitis/drug therapy , CD4-Positive T-Lymphocytes , Piperidines/pharmacology , Colitis, Ulcerative/drug therapy
4.
Antioxidants (Basel) ; 11(12)2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36552634

ABSTRACT

BACKGROUND: CD4+ T cells critically contribute to the initiation and perturbation of inflammation. When CD4+ T cells enter inflamed tissues, they adapt to hypoxia and oxidative stress conditions, and to a reduction in nutrients. We aimed to investigate how this distinct environment regulates T cell responses within the inflamed joints of patients with childhood rheumatism (JIA) by analyzing the behavior of NRF2-the key regulator of the anti-oxidative stress response-and its signaling pathways. METHODS: Flow cytometry and quantitative RT-PCR were used to perform metabolic profiling of T cells and to measure the production of inflammatory cytokines. Loss of function analyses were carried out by means of siRNA transfection experiments. NRF2 activation was induced by treatment with 4-octyl-Itaconate (4-OI). RESULTS: Flow cytometry analyses revealed a high metabolic status in CD4+ T cells taken from synovial fluid (SF) with greater mitochondrial mass, and increased glucose and fatty acid uptake. This resulted in a heightened oxidative status of SF CD4+ T cells. Despite raised ROS levels, expression of NRF2 and its target gene NQO1 were lower in CD4+ T cells from SF than in those from blood. Indeed, NRF2 activation of CD4+ T cells downregulated oxidative stress markers, altered the metabolic phenotype and reduced secretion of IFN-γ. CONCLUSION: NRF2 could be a potential regulator in CD4+ T cells during chronic inflammation and could instigate a drift toward disease progression or regression, depending on the inflammatory environment.

5.
Rheumatology (Oxford) ; 61(6): 2694-2703, 2022 05 30.
Article in English | MEDLINE | ID: mdl-34559194

ABSTRACT

OBJECTIVE: JIA is a chronic inflammatory disease of unknown origin. The regulation of inflammatory processes involves multiple cellular steps including mRNA transcription and translation. Different miRNAs control these processes tightly. We aimed to determine the roles of specific miRNAs within JIA pathogenesis. METHODS: We performed a global miRNA expression analysis in parallel in cells from the arthritic joint and peripheral blood of oligoarticular JIA patients and healthy controls. Quantitative RT-PCR analysis was used to verify expression of miRNA in T cells. Ex vivo experiments and flow cytometric analyses were used to analyse proliferation and redox metabolism. RESULTS: Global miRNA expression analysis demonstrated a different composition of miRNA expression at the site of inflammation compared with peripheral blood. Bioinformatic analysis of predicted miRNA target genes suggest a huge overrepresentation of genes involved in metabolic and oxidative stress pathways in the inflamed joint. Despite enhanced reactive oxygen species (ROS) levels within the local inflammatory milieu, JIA T cells are hyperproliferative and reveal an overexpression of miR-23a, which is an inhibitor of Peptidyl-prolyl isomerase F (PPIF), the regulator of mitochondrial ROS escape. Mitochondrial ROS escape is diminished in JIA T cells, resulting in their prolonged survival. CONCLUSION: Our data suggest that miRNA-dependent mitochondrial ROS shuttling might be a mechanism that contributes to T cell regulation in JIA at the site of inflammation.


Subject(s)
Arthritis, Juvenile , MicroRNAs , Humans , Inflammation/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , T-Lymphocytes/metabolism
7.
Nat Commun ; 12(1): 4077, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34210970

ABSTRACT

Emerging data demonstrate that the activity of immune cells can be modulated by microbial molecules. Here, we show that the short-chain fatty acids (SCFAs) pentanoate and butyrate enhance the anti-tumor activity of cytotoxic T lymphocytes (CTLs) and chimeric antigen receptor (CAR) T cells through metabolic and epigenetic reprograming. We show that in vitro treatment of CTLs and CAR T cells with pentanoate and butyrate increases the function of mTOR as a central cellular metabolic sensor, and inhibits class I histone deacetylase activity. This reprogramming results in elevated production of effector molecules such as CD25, IFN-γ and TNF-α, and significantly enhances the anti-tumor activity of antigen-specific CTLs and ROR1-targeting CAR T cells in syngeneic murine melanoma and pancreatic cancer models. Our data shed light onto microbial molecules that may be used for enhancing cellular anti-tumor immunity. Collectively, we identify pentanoate and butyrate as two SCFAs with therapeutic utility in the context of cellular cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Fatty Acids, Volatile/metabolism , Immunologic Factors/metabolism , Immunotherapy, Adoptive/methods , Microbiota/physiology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Butyrates/metabolism , Cell Line, Tumor , Cytokines/metabolism , Female , Immunotherapy , Interferon-gamma , Interleukin-2 Receptor alpha Subunit , Megasphaera , Melanoma/metabolism , Mice , Mice, Inbred C57BL , Peptide Fragments , Receptor Tyrosine Kinase-like Orphan Receptors , Receptors, G-Protein-Coupled/genetics , Tumor Necrosis Factor-alpha
8.
J Clin Invest ; 131(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-33945503

ABSTRACT

BACKGROUNDDeciphering the function of the many genes previously classified as uncharacterized open reading frame (ORF) would complete our understanding of a cell's function and its pathophysiology.METHODSWhole-exome sequencing, yeast 2-hybrid and transcriptome analyses, and molecular characterization were performed in this study to uncover the function of the C2orf69 gene.RESULTSWe identified loss-of-function mutations in the uncharacterized C2orf69 gene in 8 individuals with brain abnormalities involving hypomyelination and microcephaly, liver dysfunction, and recurrent autoinflammation. C2orf69 contains an N-terminal signal peptide that is required and sufficient for mitochondrial localization. Consistent with mitochondrial dysfunction, the patients showed signs of respiratory chain defects, and a CRISPR/Cas9-KO cell model of C2orf69 had similar respiratory chain defects. Patient-derived cells revealed alterations in immunological signaling pathways. Deposits of periodic acid-Schiff-positive (PAS-positive) material in tissues from affected individuals, together with decreased glycogen branching enzyme 1 (GBE1) activity, indicated an additional impact of C2orf69 on glycogen metabolism.CONCLUSIONSOur study identifies C2orf69 as an important regulator of human mitochondrial function and suggests that this gene has additional influence on other metabolic pathways.


Subject(s)
Glycogen/metabolism , Loss of Function Mutation , Microcephaly/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Open Reading Frames , Animals , Cell Line , Glycogen/genetics , Glycogen Debranching Enzyme System/genetics , Glycogen Debranching Enzyme System/metabolism , Humans , Mice , Mice, Knockout , Microcephaly/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics
9.
Front Immunol ; 12: 633845, 2021.
Article in English | MEDLINE | ID: mdl-33968025

ABSTRACT

Oxidative stress is a major component of cellular damage in T cells from patients with systemic lupus erythematosus (SLE) resulting amongst others in the generation of pathogenic Th17 cells. The NRF2/Keap1 pathway is the most important antioxidant system protecting cells from damage due to oxidative stress. Activation of NRF2 therefore seems to represent a putative therapeutic target in SLE, which is nevertheless challenged by several findings suggesting tissue and cell specific differences in the effect of NRF2 expression. This review focusses on the current understanding of oxidative stress in SLE T cells and its pathophysiologic and therapeutic implications.


Subject(s)
Lupus Erythematosus, Systemic/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Th17 Cells/metabolism , Animals , Antioxidants/therapeutic use , Forkhead Transcription Factors/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/immunology , Oxidation-Reduction , Oxidative Stress/drug effects , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Th17 Cells/drug effects , Th17 Cells/immunology
10.
Int J Mol Sci ; 22(8)2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33924101

ABSTRACT

MΦ differentiate from circulating monocytes (Mo). The reduced ability of neonatal Mo to undergo apoptosis after E. coli infection (phagocytosis-induced cell death (PICD)) could contribute to sustained inflammatory processes. The objective of our study was to investigate whether immune metabolism in Mo can be modified to gain access to pro-apoptotic signaling. To this end, we supplemented Mo from neonates and from adults with the branched amino acid leucine. In neonatal Mo, we observed increased energy production via oxidative phosphorylation (Oxphos) after E. coli infection via Seahorse assay. Leucine did not change phagocytic properties. In neonatal Mo, we detected temporal activation of the AKT and mTOR pathways, accompanied with subsequent activation of downstream targets S6 Kinase (S6K) and S6. FACS analyses showed that once mTOR activation was terminated, the level of anti-apoptotic BCL-2 family proteins (BCL-2; BCL-XL) decreased. Release of cytochrome C and cleavage of caspase-3 indicated involvement of the intrinsic apoptotic pathway. Concomitantly, the PICD of neonatal Mo was initiated, as detected by hypodiploid DNA. This process was sensitive to rapamycin and metformin, suggesting a functional link between AKT, mTOR and the control of intrinsic apoptotic signaling. These features were unique to neonatal Mo and could not be observed in adult Mo. Supplementation with leucine therefore could be beneficial to reduce sustained inflammation in septic neonates.


Subject(s)
Cell Death , Escherichia coli Infections/metabolism , Escherichia coli , Leucine/metabolism , Monocytes/physiology , Phagocytosis , Signal Transduction , TOR Serine-Threonine Kinases , Apoptosis , Dietary Supplements , Energy Metabolism , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Glucose/metabolism , Humans , Leucine/administration & dosage , Phagocytosis/immunology , TOR Serine-Threonine Kinases/metabolism
11.
Sci Rep ; 10(1): 624, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31953452

ABSTRACT

Neonatal sepsis is accompanied by impaired apoptotic depletion of monocytes and macrophages (MΦ), aberrant cytokine production, impaired cell metabolism, and sustained inflammation. Macrophage-colony stimulating factor (M-CSF) triggers the differentiation from monocytes into MΦ (MΦ-0). Interleukin-10 (IL10) and Interferon-gamma (IFNy) further differentiate MΦ subpopulations, the anti-inflammatory MΦ-IL10 and the pro-inflammatory MΦ-IFNy subtype. We previously have shown significant differences between adult (PBMΦ) and cord blood (CBMΦ) in the metabolism of all subtypes. To test the hypothesis whether the competence to differentiate monocytes into MΦ-0 and to polarise into MΦ-IFNy and MΦ-IL10 was diminished in CBMΦ as compared to PBMΦ, we polarised monocytes by cultivation with M-CSF for 72 h, followed by stimulation with IFNy or IL10, for 48 h. After flow cytometry based immunotyping, we tested four functions: Phagocytosis of GFP-E. coli, uptake of erythrocytes, T-cell proliferation, induction of regulatory T-cells as well as phosphorylation analysis of AKT and STAT1/STAT3. Phosphorylation of STAT-1 and STAT-3, obligatory to differentiate into MΦ-IFNγ, MΦ-0 and MΦ-IL10, was found to be aberrant in CBMΦ. Whereas infected MΦ-0 showed identical phagocytic indices and intracellular degradation, TLR4-expression, NFkB up-regulation, IL10-, IL6-, and TNFα production of CBMΦ-0 were reduced. In addition, the capacity to bind aged erythrocytes and the consecutive IL10 production was lower in CBMΦ-IL10. Polarised PBMΦ-IFNy and PBMΦ-IL10 expressed higher levels of co-stimulatory receptors (CD80, CD86), had a higher capacity to stimulate T-cells and induced higher amounts of regulatory T-cells (all p < 0.05 vs. corresponding CBMΦ). Hypoxia-inducible-factor-1α (HIF-1α) was stronger expressed in CBMΦ-IFNy and upregulated in infected CBMΦ-0, whereas heme-oxygenase 1 (HO-1) expression was similar to adult PBMΦ. Neonatal MΦ-0, MΦ-IFNy and MΦ-IL10 polarisation is impaired with respect to phenotype and functions tested which may contribute to sustained inflammation in neonatal sepsis.


Subject(s)
Interferon-gamma/metabolism , Interleukin-10/metabolism , Macrophages/cytology , Monocytes/cytology , Sepsis/immunology , Adult , Cell Polarity , Cells, Cultured , Female , Flow Cytometry , Gene Expression Regulation , Humans , Infant, Newborn , Macrophage Colony-Stimulating Factor/pharmacology , Macrophages/immunology , Male , Models, Biological , Monocytes/immunology
12.
Allergy ; 75(3): 603-615, 2020 03.
Article in English | MEDLINE | ID: mdl-31494944

ABSTRACT

BACKGROUND: Allergic diseases and especially allergic asthma are widespread diseases with high prevalence in childhood, but also in adults. Acid sphingomyelinase (ASM) is a key regulator of the sphingolipid pathway. Previous studies defined the association of ASM with the pathogenesis of TH 1-directed lung diseases like cystic fibrosis and acute lung injury. Here, we define the role of ASM in TH 2-regulated allergic bronchial asthma. METHODS: To determine the role of Asm under baseline conditions, wild-type (WT) and Asm-/- mice were ventilated with a flexiVent setup and bronchial hyperresponsiveness was determined using acetylcholine. Flow cytometry and cytokine measurements in bronchoalveolar lavage fluid and lung tissue were followed by in vitro TH 2 differentiations with cells from WT and Asm-/- mice and blockade of Asm with amitriptyline. As proof of principle, we conducted an ovalbumin-induced model of asthma in WT- and Asm-/-  mice. RESULTS: At baseline, Asm-/- mice showed better lung mechanics, but unaltered bronchial hyperresponsiveness. Higher numbers of Asm-/- T cells in bronchoalveolar lavage fluid released lower levels of IL-4 and IL-5, and these results were paralleled by decreased production of typical TH 2 cytokines in Asm-/- T lymphocytes in vitro. This phenotype could be imitated by incubation of T cells with amitriptyline. In the ovalbumin asthma model, Asm-/- animals were protected from high disease activity and showed better lung functions and lower levels of eosinophils and TH 2 cytokines. CONCLUSION: Asm deficiency could induce higher numbers of TH 2 cells in the lung, but those cells release decreased TH 2 cytokine levels. Hereby, Asm-/- animals are protected from bronchial asthma, which possibly offers novel therapeutic strategies, for example, with ASM blockade.


Subject(s)
Asthma , Bronchial Hyperreactivity , Animals , Bronchoalveolar Lavage Fluid , Cytokines , Disease Models, Animal , Lung , Mice , Mice, Inbred BALB C , Ovalbumin , Sphingomyelin Phosphodiesterase/genetics , Th2 Cells
13.
Kidney Int ; 97(2): 289-303, 2020 02.
Article in English | MEDLINE | ID: mdl-31882173

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease and lupus nephritis is a major risk factor for morbidity and mortality. Notch-3 signaling induced by membrane-bound or soluble ligands such as YB-1 constitutes an evolutionarily conserved pathway that determines major decisions in cell fate. Mass spectrometry of extracellular YB-1 in sera from patients with SLE and lupus-prone mice revealed specific post-translational guanidinylation of two lysine residues within the highly conserved cold-shock domain of YB-1 (YB-1-G). These modifications highly correlated with SLE disease activity, especially in patients with lupus nephritis and resulted in enhanced activation of Notch-3 signaling in T lymphocytes. The importance of YB-1:Notch-3 interaction in T cells was further evidenced by increased interleukin (Il)10 expression following YB-1-G stimulation and detection of both, YB-1-G and Notch-3, in kidneys of MRL.lpr mice by mass spectrometry imaging. Notch-3 expression and activation was significantly up-regulated in kidneys of 20-week-old MRL.lpr mice. Notably, lupus-prone mice with constitutional Notch-3 depletion (B6.Faslpr/lprNotch3-/-) exhibited an aggravated lupus phenotype with significantly increased mortality, enlarged lymphoid organs and aggravated nephritis. Additionally, these mice displayed fewer regulatory T cells and reduced amounts of anti-inflammatory IL-10. Thus, our results indicate that the YB-1:Notch-3 axis exerts protective effects in SLE and that Notch-3 deficiency exacerbates the SLE phenotype.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Receptor, Notch3/metabolism , Transcription Factors/metabolism , Animals , Humans , Lupus Erythematosus, Systemic/complications , Mice , Mice, Inbred MRL lpr , Signal Transduction , T-Lymphocytes, Regulatory
14.
Eur J Immunol ; 50(4): 515-524, 2020 04.
Article in English | MEDLINE | ID: mdl-31840803

ABSTRACT

The transcription factor Nrf2 regulates oxidative stress responses. However, the specific function of Nrf2 in Tregs, the central regulators of immune homeostasis, is unclear. Here, we report an unexpected but important role of Nrf2 in Tregs. Nrf2 expression driven by Foxp3 specific deletion of Keap1 resulted in an autoinflammatory phenotype with enhanced effector T cell activation and immune cell infiltrates in the lung. While early postnatal death of mice with Foxp3 specific deletion of Keap1 was most probably due to ectopic Foxp3cre expression and subsequent Keap1 deletion in epithelial cells, bone marrow chimeras suggest that Nrf2 activation intrinsically in Tregs contributes to a loss of Treg cells and diminished peripheral tolerance. Moreover, Nrf2 activation was associated with a loss of Foxp3 expression, but an enhanced glucose uptake and mTOR activity in Tregs, thus mimicking a metabolic phenotype that is associated with impaired lineage stability and cell functioning.


Subject(s)
Inflammation/immunology , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity , Chimera , Forkhead Transcription Factors/metabolism , Homeostasis , Immune Tolerance , Immunomodulation , Kelch-Like ECH-Associated Protein 1/genetics , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , TOR Serine-Threonine Kinases/metabolism
15.
Nat Commun ; 10(1): 1685, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30976008

ABSTRACT

Neonatal sepsis is characterized by hyperinflammation causing enhanced morbidity and mortality compared to adults. This suggests differences in the response towards invading threats. Here we investigate activated cord blood macrophages (CBMΦ) in comparison to adult macrophages (PBMΦ), indicating incomplete interferon gamma (IFN-γ) and interleukin 10 (IL-10)-induced activation of CBMΦ. CBMΦ show reduced expression of phagocytosis receptors and cytokine expression in addition to altered energy metabolism. In particular, IFN-γ as well as IL-10-activated CBMΦ completely fail to increase glycolysis and furthermore show reduced activation of the mTOR pathway, which is important for survival in sepsis. MTOR inhibition by rapamycin equalizes cytokine production in CBMΦ and PBMΦ. Finally, incubation of PBMΦ with cord blood serum or S100A8/A9, which is highly expressed in neonates, suppresses mTOR activation, prevents glycolysis and the expression of an PBMΦ phenotype. Thus, a metabolic alteration is apparent in CBMΦ, which might be dependent on S100A8/A9 expression.


Subject(s)
Cytotoxicity, Immunologic , Energy Metabolism/immunology , Macrophages/metabolism , Adult , Age Factors , Calgranulin A/immunology , Calgranulin A/metabolism , Calgranulin B/immunology , Calgranulin B/metabolism , Cell Differentiation/immunology , Cells, Cultured , Fetal Blood/cytology , Glycolysis/immunology , Healthy Volunteers , Humans , Infant, Newborn , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Macrophages/immunology , Primary Cell Culture , Sepsis/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
16.
Front Immunol ; 9: 2499, 2018.
Article in English | MEDLINE | ID: mdl-30425715

ABSTRACT

Reactive oxygen species (ROS) molecules are implicated in signal transduction pathways and thereby control a range of biological activities. Immune cells are constantly confronted with ROS molecules under both physiologic and pathogenic conditions. Myeloid-derived suppressor cells (MDSCs) are immunosuppressive, immature myeloid cells and serve as major regulators of pathogenic and inflammatory immune responses. In addition to their own release of ROS, MDSCs often arise in oxidative-stress prone environments such as in tumors or during inflammation and infection. This evidently close relationship between MDSCs and ROS prompted us to summarize what is currently known about ROS signaling within MDSCs and to elucidate how MDSCs use ROS to modulate other immune cells. ROS not only activate anti-oxidative pathways but also induce transcriptional programs that regulate the fate and function of MDSCs. Furthermore, MDSCs release ROS molecules as part of a major mechanism to suppress T cell responses. Targeting redox-regulation of MDSCs thus presents a promising approach to cancer therapy and the role of redox-signaling in MDSCs in other disease states such as infection, inflammation and autoimmunity would appear to be well worth investigating.


Subject(s)
Inflammation/immunology , Myeloid-Derived Suppressor Cells/immunology , Oxidative Stress , Reactive Oxygen Species/metabolism , Animals , Cellular Reprogramming , Humans , Immune Tolerance , Oxidation-Reduction , Signal Transduction
17.
BMC Pulm Med ; 18(1): 174, 2018 Nov 22.
Article in English | MEDLINE | ID: mdl-30466430

ABSTRACT

BACKGROUND: The acute respiratory distress syndrome (ARDS) is a serious disease in critically ill patients that is characterized by pulmonary dysfunctions, hypoxemia and significant mortality. Patients with immunodeficiency (e.g. SCID with T and B cell deficiency) are particularly susceptible to the development of severe ARDS. However, the role of T cells on pulmonary dysfunctions in immune-competent patients with ARDS is only incompletely understood. METHODS: Wild-type (wt) and RAG2-/- mice (lymphocyte deficient) received intratracheal instillations of LPS (4 mg/kg) or saline. On day 1, 4 and 10 lung mechanics and bronchial hyperresponsiveness towards acetylcholine were measured with the flexiVent ventilation set-up. The bronchoalveolar lavage fluid (BALF) was examined for leukocytes (FACS analysis) and pro-inflammatory cytokines (ELISA). RESULTS: In wt mice, lung mechanics, body weight and body temperature deteriorated in the LPS-group during the early phase (up to d4); these alterations were accompanied by increased leukocyte numbers and inflammatory cytokine levels in the BALF. During the late phase (day 10), both lung mechanics and the cell/cytokine homeostasis recovered in LPS-treated wt mice. RAG2-/- mice experienced changes in body weight, lung mechanics, BAL neutrophil numbers, BAL inflammatory cytokines levels that were comparable to wt mice. CONCLUSION: Following LPS instillation, lung mechanics deteriorate within the first 4 days and recover towards day 10. This response is not altered by the lack of T lymphocytes suggesting that T cells play only a minor role for the initiation, propagation or recovery of LPS-induced lung dysfunctions or function of T lymphocytes can be compensated by other immune cells, such as alveolar macrophages.


Subject(s)
Acute Lung Injury/immunology , Cytokines/metabolism , Respiratory Distress Syndrome/immunology , T-Lymphocytes/immunology , Acute Lung Injury/chemically induced , Animals , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Female , Lipopolysaccharides , Lung/physiopathology , Macrophages, Alveolar , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Respiratory Distress Syndrome/chemically induced
18.
Front Immunol ; 9: 1552, 2018.
Article in English | MEDLINE | ID: mdl-30034396

ABSTRACT

Arising in inflammatory conditions, myeloid-derived suppressor cells (MDSCs) are constantly confronted with intracellular and extracellular reactive oxygen species molecules and oxidative stress. Generating mice with a constitutive activation of Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) we show a pivotal role of the antioxidant stress defense for development of these immune-modulatory cells. These mice are characterized by a massive increase of splenic CD11b+Gr-1+ cells, which exhibit typical suppressive characteristics of MDSCs. Whole transcriptome analysis revealed Nrf2-dependent activation of cell cycle and metabolic pathways, which resemble pathways in CD11b+Gr-1+ MDSCs expanded by in vivo LPS exposure. Constitutive Nrf2 activation thereby regulates activation and balance between glycolysis and mitochondrial metabolism and hence expansion of highly suppressive MDSCs, which mediate protection in LPS-induced sepsis. Our study establishes Nrf2 as key regulator of MDSCs and acquired tolerance against LPS-induced sepsis.

19.
J Immunol Res ; 2018: 8947230, 2018.
Article in English | MEDLINE | ID: mdl-29854847

ABSTRACT

Dendritic cells (DCs) are crucial for the balance between immune response and tolerance, but the molecular mechanism regulating development, differentiation, and homeostasis are poorly understood. The transcriptional activator CREB is involved in regulating different cells of the innate and adaptive immune system and is a transcriptional regulator of development, survival, activation, or proliferation in macrophages, dendritic cells, B cells, and T cells. To directly examine the role of CREB in the regulation of DCs, the CREB gene was targeted for deletion with a CD11c-cre transgene. The deletion of CREB in CD11c+ cells did not involve any developmental or systemic defects within DC populations. However, CREB deficiency in CD11c+ cells reduced germinal center (GC) B cells in steady state, and immunization with NP-CGG resulted in a reduced formation of GCs, paralleled by the reduced production of IgGs in sera of immunized mice. In conclusion, we demonstrate that CREB expression in CD11c+ cells enhances germinal center responses, most likely by altering DC function, which might have implications for autoimmune diseases that are associated with dysregulated GC responses.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocytes/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Dendritic Cells/immunology , Germinal Center/immunology , Animals , Antigen Presentation , CD11c Antigen/metabolism , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Immunization , Immunoglobulin G/blood , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
Pediatr Rheumatol Online J ; 16(1): 39, 2018 Jun 20.
Article in English | MEDLINE | ID: mdl-29925386

ABSTRACT

BACKGROUND: Inflammatory effector T cells trigger inflammation despite increased numbers of Treg cells in the synovial joint of patients suffering from juvenile idiopathic arthritis (JIA). The cAMP response element (CREM)α is known to play a major role in regulation of T cells in SLE, colitis, and EAE. However, its role in regulation of effector T cells within the inflammatory joint is unknown. METHODS: CREM expression was analyzed in synovial fluid cells from oligoarticular JIA patients by flow cytometry. Peripheral blood mononuclear cells were incubated with synovial fluid and analyzed in the presence and absence of CREM using siRNA experiments for T cell phenotypes. To validate the role of CREM in vivo, ovalbumin-induced T cell dependent arthritis experiments were performed. RESULTS: CREM is highly expressed in synovial fluid T cells and its expression can be induced by treating healthy control PBMCs with synovial fluid. Specifically, CREM is more abundant in CD161+ subsets, than CD161- subsets, of T cells and contributes to cytokine expression by these cells. Finally, development of ovalbumin-induced experimental arthritis is ameliorated in mice with adoptively transferred CREM-/- T cells. CONCLUSION: In conclusion, our study reveals that beyond its role in SLE T cells CREM also drives an inflammatory phenotype of T cells in JIA.


Subject(s)
Arthritis, Juvenile/metabolism , Cyclic AMP Response Element Modulator/metabolism , Synovial Fluid/metabolism , T-Lymphocytes/metabolism , Animals , Cell Culture Techniques , Flow Cytometry/methods , Humans , Joints/pathology , Mice , Phenotype , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...