Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Clin Immunol ; 257: 109817, 2023 12.
Article in English | MEDLINE | ID: mdl-37925120

ABSTRACT

A subset of autoimmune diseases is characterized by predominant pathogenic IgG4 autoantibodies (IgG4-AID). Why IgG4 predominates in these disorders is unknown. We hypothesized that dysregulated B cell maturation or aberrant class switching causes overrepresentation of IgG4+ B cells and plasma cells. Therefore, we compared the B cell compartment of patients from four different IgG4-AID with two IgG1-3-AID and healthy donors, using flow cytometry. Relative subset abundance at all maturation stages was normal, except for a, possibly treatment-related, reduction in immature and naïve CD5+ cells. IgG4+ B cell and plasma cell numbers were normal in IgG4-AID patients, however they had a (sub)class-independent 8-fold increase in circulating CD20-CD138+ cells. No autoreactivity was found in this subset. These results argue against aberrant B cell development and rather suggest the autoantibody subclass predominance to be antigen-driven. The similarities between IgG4-AID suggest that, despite displaying variable clinical phenotypes, they share a similar underlying immune profile.


Subject(s)
Autoantibodies , Autoimmune Diseases , Humans , Immunoglobulin Class Switching , Immunoglobulin G , B-Lymphocytes
2.
Rheumatology (Oxford) ; 61(6): 2682-2693, 2022 05 30.
Article in English | MEDLINE | ID: mdl-34559222

ABSTRACT

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


Subject(s)
Platelet Factor 4/metabolism , Reactive Oxygen Species/metabolism , Scleroderma, Systemic , Toll-Like Receptor 9 , Basic Helix-Loop-Helix Transcription Factors/metabolism , Dendritic Cells/metabolism , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit
3.
Int J Mol Sci ; 20(17)2019 Aug 25.
Article in English | MEDLINE | ID: mdl-31450682

ABSTRACT

Cells are exposed to reactive oxygen species (ROS) as a by-product of mitochondrial metabolism, especially under hypoxia. ROS are also enzymatically generated at the plasma membrane during inflammation. Radicals cause cellular damage leading to cell death, as they react indiscriminately with surrounding lipids, proteins, and nucleotides. However, ROS are also important for many physiological processes, including signaling, pathogen killing and chemotaxis. The sensitivity of cells to ROS therefore likely depends on the subcellular location of ROS production, but how this affects cell viability is poorly understood. As ROS generation consumes oxygen, and hypoxia-mediated signaling upregulates expression of antioxidant transcription factor Nrf2, it is difficult to discern hypoxic from radical stress. In this study, we developed an optogenetic toolbox for organelle-specific generation of ROS using the photosensitizer protein SuperNova which produces superoxide anion upon excitation with 590 nm light. We fused SuperNova to organelle specific localization signals to induce ROS with high precision. Selective ROS production did not affect cell viability in most organelles except for the nucleus. SuperNova is a promising tool to induce locally targeted ROS production, opening up new possibilities to investigate processes and organelles that are affected by localized ROS production.


Subject(s)
Cell Nucleus/metabolism , Free Radicals/metabolism , Organelles/metabolism , Oxidative Stress , Animals , Biomarkers , COS Cells , Cell Death , Cell Nucleus/genetics , Chlorocebus aethiops , DNA Damage , Reactive Oxygen Species/metabolism
4.
Front Immunol ; 10: 1216, 2019.
Article in English | MEDLINE | ID: mdl-31191556

ABSTRACT

Neutrophils kill ingested pathogens by the so-called oxidative burst, where reactive oxygen species (ROS) are produced in the lumen of phagosomes at very high rates (mM/s), although these rates can only be maintained for a short period (minutes). In contrast, dendritic cells produce ROS at much lower rates, but they can sustain production for much longer after pathogen uptake (hours). It is becoming increasingly clear that this slow but prolonged ROS production is essential for antigen cross-presentation to activate cytolytic T cells, and for shaping the repertoire of antigen fragments for presentation to helper T cells. However, despite this importance of ROS production by dendritic cells for activation of the adaptive immune system, their actual ROS production rates have never been quantified. Here, we quantified ROS production in human monocyte-derived dendritic cells by measuring the oxygen consumption rate during phagocytosis. Although a large variation in oxygen consumption and phagocytic capacity was present among individuals and cells, we estimate a ROS production rate of on average ~0.5 mM/s per phagosome. Quantitative microscopy approaches showed that ROS is produced within minutes after pathogen encounter at the nascent phagocytic cup. H2DCFDA measurements revealed that ROS production is sustained for at least ~10 h after uptake. While ROS are produced by dendritic cells at an about 10-fold lower rate than by neutrophils, the net total ROS production is approximately similar. These are the first quantitative estimates of ROS production by a cell capable of antigen cross-presentation. Our findings provide a quantitative insight in how ROS affect dendritic cell function.


Subject(s)
Dendritic Cells/metabolism , Monocytes/cytology , Phagosomes/metabolism , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Cross-Priming , Dendritic Cells/drug effects , Dendritic Cells/ultrastructure , Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Humans , Kinetics , Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Mice , NADPH Oxidase 2/metabolism , Oxygen Consumption , Phosphoproteins/metabolism , RAW 264.7 Cells , Superoxides/metabolism , Zymosan/pharmacology
5.
Oncotarget ; 10(8): 883-896, 2019 Jan 25.
Article in English | MEDLINE | ID: mdl-30783517

ABSTRACT

Solid tumors grow at a high speed leading to insufficient blood supply to tumor cells. This makes the tumor hypoxic, resulting in the Warburg effect and an increased generation of reactive oxygen species (ROS). Hypoxia and ROS affect immune cells in the tumor micro-environment, thereby affecting their immune function. Here, we review the known effects of hypoxia and ROS on the function and physiology of dendritic cells (DCs). DCs can (cross-)present tumor antigen to activate naive T cells, which play a pivotal role in anti-tumor immunity. ROS might enter DCs via aquaporins in the plasma membrane, diffusion across the plasma membrane or via extracellular vesicles (EVs) released by tumor cells. Hypoxia and ROS exert complex effects on DCs, and can both inhibit and activate maturation of immature DCs. Furthermore, ROS transferred by EVs and/or produced by the DC can both promote antigen (cross-)presentation through phagosomal alkalinization, which preserves antigens by inhibiting proteases, and by direct oxidative modification of proteases. Hypoxia leads to a more migratory and inflammatory DC phenotype. Lastly, hypoxia alters DCs to shift the T- cell response towards a tumor suppressive Th17 phenotype. From numerous studies, the concept is emerging that hypoxia and ROS are mutually dependent effectors on DC function in the tumor micro-environment. Understanding their precise roles and interplay is important given that an adaptive immune response is required to clear tumor cells.

6.
Biosci Rep ; 38(6)2018 12 21.
Article in English | MEDLINE | ID: mdl-30463908

ABSTRACT

Dendritic cells (DCs) constantly sample peripheral tissues for antigens, which are subsequently ingested to derive peptides for presentation to T cells in lymph nodes. To do so, DCs have to traverse many different tissues with varying oxygen tensions. Additionally, DCs are often exposed to low oxygen tensions in tumors, where vascularization is lacking, as well as in inflammatory foci, where oxygen is rapidly consumed by inflammatory cells during the respiratory burst. DCs respond to oxygen levels to tailor immune responses to such low-oxygen environments. In the present study, we identified a mechanism of hypoxia-mediated potentiation of release of tumor necrosis factor α (TNF-α), a pro-inflammatory cytokine with important roles in both anti-cancer immunity and autoimmune disease. We show in human monocyte-derived DCs (moDCs) that this potentiation is controlled exclusively via the p38/mitogen-activated protein kinase (MAPK) pathway. We identified MAPK kinase kinase 8 (MAP3K8) as a target gene of hypoxia-induced factor (HIF), a transcription factor controlled by oxygen tension, upstream of the p38/MAPK pathway. Hypoxia increased expression of MAP3K8 concomitant with the potentiation of TNF-α secretion. This potentiation was no longer observed upon siRNA silencing of MAP3K8 or with a small molecule inhibitor of this kinase, and this also decreased p38/MAPK phosphorylation. However, expression of DC maturation markers CD83, CD86, and HLA-DR were not changed by hypoxia. Since DCs play an important role in controlling T-cell activation and differentiation, our results provide novel insight in understanding T-cell responses in inflammation, cancer, autoimmune disease and other diseases where hypoxia is involved.


Subject(s)
Dendritic Cells/immunology , Hypoxia/immunology , Inflammation/immunology , MAP Kinase Kinase Kinases/immunology , Proto-Oncogene Proteins/immunology , Tumor Necrosis Factor-alpha/immunology , Cell Hypoxia , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Hypoxia/genetics , Inflammation/genetics , MAP Kinase Kinase Kinases/genetics , Monocytes/cytology , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Toll-Like Receptor 4/immunology , Up-Regulation
7.
Sci Rep ; 7(1): 6889, 2017 07 31.
Article in English | MEDLINE | ID: mdl-28761087

ABSTRACT

Ethylene is a major plant hormone mediating developmental processes and stress responses to stimuli such as infection. We show here that ethylene is also produced during systemic inflammation in humans and is released in exhaled breath. Traces of ethylene were detected by laser spectroscopy both in vitro in isolated blood leukocytes exposed to bacterial lipopolysaccharide (LPS) as well as in vivo following LPS administration in healthy volunteers. Exposure to LPS triggers formation of ethylene as a product of lipid peroxidation induced by the respiratory burst. In humans, ethylene was detected prior to the increase of blood levels of inflammatory cytokines and stress-related hormones. Our results highlight that ethylene release is an early and integral component of in vivo lipid peroxidation with important clinical implications as a breath biomarker of bacterial infection.


Subject(s)
Ethylenes/analysis , Inflammation/metabolism , Leukocytes/metabolism , Lipopolysaccharides/adverse effects , Adult , Biomarkers/analysis , Breath Tests , Cytokines/metabolism , Healthy Volunteers , Humans , Inflammation/chemically induced , Inflammation/immunology , Leukocytes/drug effects , Lipid Peroxidation , Lipopolysaccharides/pharmacology , Male , Respiratory Burst , Young Adult
8.
Eur J Cell Biol ; 96(7): 705-714, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28688576

ABSTRACT

Cross-presentation of foreign antigen in major histocompatibility complex (MHC) class I by dendritic cells (DCs) requires activation of the NADPH-oxidase NOX2 complex. We recently showed that NOX2 is recruited to phagosomes by the SNARE protein VAMP8 where NOX2-produced reactive oxygen species (ROS) cause lipid oxidation and membrane disruption, promoting antigen translocation into the cytosol for cross-presentation. In this study, we extend these findings by showing that VAMP8 is also involved in NOX2 trafficking to endosomes. Moreover, we demonstrate in both human and mouse DCs that absence of VAMP8 leads to decreased ROS production, lipid peroxidation and antigen translocation, and that this impairs cross-presentation. In contrast, knockdown of VAMP8 did not affect recruitment of MHC class I and the transporter associated with antigen processing 1 (TAP1) to phagosomes, although surface levels of MHC class I were reduced. Thus, in addition to a secretory role, VAMP8-mediates trafficking of NOX2 to endosomes and phagosomes and this promotes induction of cytolytic T cell immune responses.


Subject(s)
Antigen Presentation/genetics , Dendritic Cells/immunology , NADPH Oxidase 2/genetics , R-SNARE Proteins/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 2/immunology , Animals , Antigen Presentation/immunology , Cell Membrane/genetics , Cell Membrane/immunology , Endosomes/genetics , Endosomes/immunology , Genes, MHC Class I/immunology , Humans , Lipid Peroxidation , Mice , NADPH Oxidase 2/immunology , Phagosomes/genetics , Phagosomes/immunology , R-SNARE Proteins/immunology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , T-Lymphocytes, Cytotoxic/immunology
9.
Nat Commun ; 7: 13127, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27721497

ABSTRACT

Podosomes are cytoskeletal structures crucial for cell protrusion and matrix remodelling in osteoclasts, activated endothelial cells, macrophages and dendritic cells. In these cells, hundreds of podosomes are spatially organized in diversely shaped clusters. Although we and others established individual podosomes as micron-sized mechanosensing protrusive units, the exact scope and spatiotemporal organization of podosome clustering remain elusive. By integrating a newly developed extension of Spatiotemporal Image Correlation Spectroscopy with novel image analysis, we demonstrate that F-actin, vinculin and talin exhibit directional and correlated flow patterns throughout podosome clusters. Pattern formation and magnitude depend on the cluster actomyosin machinery. Indeed, nanoscopy reveals myosin IIA-decorated actin filaments interconnecting multiple proximal podosomes. Extending well-beyond podosome nearest neighbours, the actomyosin-dependent dynamic spatial patterns reveal a previously unappreciated mesoscale connectivity throughout the podosome clusters. This directional transport and continuous redistribution of podosome components provides a mechanistic explanation of how podosome clusters function as coordinated mechanosensory area.


Subject(s)
Actomyosin/metabolism , Cytoskeleton/metabolism , Podosomes/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Cell Surface Extensions/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Models, Biological , Nonmuscle Myosin Type IIA/metabolism , Polymerization , Rheology , Talin/metabolism , Time Factors , Vinculin/metabolism
10.
Sci Rep ; 6: 22064, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26907999

ABSTRACT

Dendritic cells (DCs) present foreign antigen in major histocompatibility complex (MHC) class I molecules to cytotoxic T cells in a process called cross-presentation. An important step in this process is the release of antigen from the lumen of endosomes into the cytosol, but the mechanism of this step is still unclear. In this study, we show that reactive oxygen species (ROS) produced by the NADPH-oxidase complex NOX2 cause lipid peroxidation, a membrane disrupting chain-reaction, which in turn results in antigen leakage from endosomes. Antigen leakage and cross-presentation were inhibited by blocking ROS production or scavenging radicals and induced when using a ROS-generating photosensitizer. Endosomal antigen release was impaired in DCs from chronic granulomatous disease (CGD) patients with dysfunctional NOX2. Thus, NOX2 induces antigen release from endosomes for cross-presentation by direct oxidation of endosomal lipids. This constitutes a new cellular function for ROS in regulating immune responses against pathogens and cancer.


Subject(s)
Cross-Priming , Dendritic Cells/immunology , Granulomatous Disease, Chronic/immunology , Lipid Peroxidation/immunology , Membrane Glycoproteins/immunology , NADPH Oxidases/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen Presentation , Dendritic Cells/cytology , Dendritic Cells/drug effects , Endosomes/immunology , Endosomes/metabolism , Free Radical Scavengers/pharmacology , Gene Expression , Granulomatous Disease, Chronic/metabolism , Granulomatous Disease, Chronic/pathology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Jurkat Cells , Lipid Peroxidation/drug effects , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidases/genetics , Photosensitizing Agents/pharmacology , Primary Cell Culture , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , alpha-Tocopherol/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL