279 results on '"Schmidt-Weber, Carsten B."'
Search Results
252. Genome-Wide Gene Expression Analysis Reveals Unique Genes Signatures of Epithelial Reorganization in Primary Airway Epithelium Induced by Type-I, -II and -III Interferons.
- Author
-
Erb A, Zissler UM, Oelsner M, Chaker AM, Schmidt-Weber CB, and Jakwerth CA
- Subjects
- Epithelium metabolism, Antiviral Agents pharmacology, Gene Expression, Interferon Type I genetics, Interferon Type I metabolism, Interferon Type I pharmacology
- Abstract
Biosensors such as toll-like receptors (TLR) induce the expression of interferons (IFNs) after viral infection that are critical to the first step in cell-intrinsic host defense mechanisms. Their differential influence on epithelial integrity genes, however, remains elusive. A genome-wide gene expression biosensor chip for gene expression sensing was used to examine the effects of type-I, -II, and -III IFN stimulation on the epithelial expression profiles of primary organotypic 3D air-liquid interface airway cultures. All types of IFNs induced similar interferon-stimulated genes (ISGs): OAS1, OAS2, and IFIT2. However, they differentially induced transcription factors, epithelial modulators, and pro-inflammatory genes. Type-I IFN-induced genes were associated with cell-cell adhesion and tight junctions, while type-III IFNs promoted genes important for transepithelial transport. In contrast, type-II IFN stimulated proliferation-triggering genes associated and enhanced pro-inflammatory mediator secretion. In conclusion, with our microarray system, we provide evidence that the three IFN types exceed their antiviral ISG-response by inducing distinct remodeling processes, thereby likely strengthening the epithelial airway barrier by enhancing cross-cell-integrity (I), transepithelial transport (III) and finally reconstruction through proliferation (II).
- Published
- 2022
- Full Text
- View/download PDF
253. Role of microRNAs in type 2 diseases and allergen-specific immunotherapy.
- Author
-
Jakwerth CA, Kitzberger H, Pogorelov D, Müller A, Blank S, Schmidt-Weber CB, and Zissler UM
- Abstract
MicroRNAs (miRs) have gained scientific attention due to their importance in the pathophysiology of allergic diseases as well as their potential as biomarkers in allergen-specific treatment options. Their function as post-transcriptional regulators, controlling various cellular processes, is of high importance since any single miR can target multiple mRNAs, often within the same signalling pathway. MiRs can alter dysregulated expression of certain cellular responses and contribute to or cause, but in some cases prevent or repress, the development of various diseases. In this review article, we describe current research on the role of specific miRs in regulating immune responses in epithelial cells and specialized immune cells in response to various stimuli, in allergic diseases, and regulation in the therapeutic approach of allergen-specific immunotherapy (AIT). Despite the fact that AIT has been used successfully as a causative treatment option since more than a century, very little is known about the mechanisms of regulation and its connections with microRNAs. In order to fill this gap, this review aims to provide an overview of the current knowledge., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (© 2022 Jakwerth, Kitzberger, Pogorelov, Müller, Blank, Schmidt-Weber and Zissler.)
- Published
- 2022
- Full Text
- View/download PDF
254. Attenuation of SARS-CoV-2 replication and associated inflammation by concomitant targeting of viral and host cap 2'-O-ribose methyltransferases.
- Author
-
Bergant V, Yamada S, Grass V, Tsukamoto Y, Lavacca T, Krey K, Mühlhofer MT, Wittmann S, Ensser A, Herrmann A, Vom Hemdt A, Tomita Y, Matsuyama S, Hirokawa T, Huang Y, Piras A, Jakwerth CA, Oelsner M, Thieme S, Graf A, Krebs S, Blum H, Kümmerer BM, Stukalov A, Schmidt-Weber CB, Igarashi M, Gramberg T, Pichlmair A, and Kato H
- Subjects
- Animals, Antiviral Agents pharmacology, Inflammation drug therapy, Methyltransferases metabolism, Mice, RNA Caps metabolism, RNA, Viral genetics, Ribose, Viral Nonstructural Proteins genetics, SARS-CoV-2, COVID-19 Drug Treatment
- Abstract
The SARS-CoV-2 infection cycle is a multistage process that relies on functional interactions between the host and the pathogen. Here, we repurposed antiviral drugs against both viral and host enzymes to pharmaceutically block methylation of the viral RNA 2'-O-ribose cap needed for viral immune escape. We find that the host cap 2'-O-ribose methyltransferase MTr1 can compensate for loss of viral NSP16 methyltransferase in facilitating virus replication. Concomitant inhibition of MTr1 and NSP16 efficiently suppresses SARS-CoV-2 replication. Using in silico target-based drug screening, we identify a bispecific MTr1/NSP16 inhibitor with anti-SARS-CoV-2 activity in vitro and in vivo but with unfavorable side effects. We further show antiviral activity of inhibitors that target independent stages of the host SAM cycle providing the methyltransferase co-substrate. In particular, the adenosylhomocysteinase (AHCY) inhibitor DZNep is antiviral in in vitro, in ex vivo, and in a mouse infection model and synergizes with existing COVID-19 treatments. Moreover, DZNep exhibits a strong immunomodulatory effect curbing infection-induced hyperinflammation and reduces lung fibrosis markers ex vivo. Thus, multispecific and metabolic MTase inhibitors constitute yet unexplored treatment options against COVID-19., (© 2022 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
255. A novel monoclonal IgG1 antibody specific for Galactose-alpha-1,3-galactose questions alpha-Gal epitope expression by bacteria.
- Author
-
Kreft L, Schepers A, Hils M, Swiontek K, Flatley A, Janowski R, Mirzaei MK, Dittmar M, Chakrapani N, Desai MS, Eyerich S, Deng L, Niessing D, Fischer K, Feederle R, Blank S, Schmidt-Weber CB, Hilger C, Biedermann T, and Ohnmacht C
- Subjects
- Animals, Antibodies, Monoclonal, Bacteria, Epitopes, Humans, Mice, Galactose, Immunoglobulin G
- Abstract
The alpha-Gal epitope (α-Gal) with the determining element galactose-α1,3-galactose can lead to clinically relevant allergic reactions and rejections in xenotransplantation. These immune reactions can develop because humans are devoid of this carbohydrate due to evolutionary loss of the enzyme α1,3-galactosyltransferase (GGTA1). In addition, up to 1% of human IgG antibodies are directed against α-Gal, but the stimulus for the induction of anti-α-Gal antibodies is still unclear. Commensal bacteria have been suggested as a causal factor for this induction as α-Gal binding tools such as lectins were found to stain cultivated bacteria isolated from the intestinal tract. Currently available tools for the detection of the definite α-Gal epitope, however, are cross-reactive, or have limited affinity and, hence, offer restricted possibilities for application. In this study, we describe a novel monoclonal IgG1 antibody (27H8) specific for the α-Gal epitope. The 27H8 antibody was generated by immunization of Ggta1 knockout mice and displays a high affinity towards synthetic and naturally occurring α-Gal in various applications. Using this novel tool, we found that intestinal bacteria reported to be α-Gal positive cannot be stained with 27H8 questioning whether commensal bacteria express the native α-Gal epitope at all., Competing Interests: MSD works as a consultant and an advisory board member at Theralution GmbH, Germany. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Kreft, Schepers, Hils, Swiontek, Flatley, Janowski, Mirzaei, Dittmar, Chakrapani, Desai, Eyerich, Deng, Niessing, Fischer, Feederle, Blank, Schmidt-Weber, Hilger, Biedermann and Ohnmacht.)
- Published
- 2022
- Full Text
- View/download PDF
256. ADAM10 and ADAM17 promote SARS-CoV-2 cell entry and spike protein-mediated lung cell fusion.
- Author
-
Jocher G, Grass V, Tschirner SK, Riepler L, Breimann S, Kaya T, Oelsner M, Hamad MS, Hofmann LI, Blobel CP, Schmidt-Weber CB, Gokce O, Jakwerth CA, Trimpert J, Kimpel J, Pichlmair A, and Lichtenthaler SF
- Subjects
- ADAM10 Protein genetics, ADAM17 Protein, Amyloid Precursor Protein Secretases genetics, Angiotensin-Converting Enzyme 2, Cell Fusion, Humans, Lung, Membrane Proteins genetics, Membrane Proteins metabolism, Metalloproteases, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, Virus Internalization, COVID-19, SARS-CoV-2
- Abstract
The severe-acute-respiratory-syndrome-coronavirus-2 (SARS-CoV-2) is the causative agent of COVID-19, but host cell factors contributing to COVID-19 pathogenesis remain only partly understood. We identify the host metalloprotease ADAM17 as a facilitator of SARS-CoV-2 cell entry and the metalloprotease ADAM10 as a host factor required for lung cell syncytia formation, a hallmark of COVID-19 pathology. ADAM10 and ADAM17, which are broadly expressed in the human lung, cleave the SARS-CoV-2 spike protein (S) in vitro, indicating that ADAM10 and ADAM17 contribute to the priming of S, an essential step for viral entry and cell fusion. ADAM protease-targeted inhibitors severely impair lung cell infection by the SARS-CoV-2 variants of concern alpha, beta, delta, and omicron and also reduce SARS-CoV-2 infection of primary human lung cells in a TMPRSS2 protease-independent manner. Our study establishes ADAM10 and ADAM17 as host cell factors for viral entry and syncytia formation and defines both proteases as potential targets for antiviral drug development., (© 2022 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2022
- Full Text
- View/download PDF
257. Lung Epithelial CYP1 Activity Regulates Aryl Hydrocarbon Receptor Dependent Allergic Airway Inflammation.
- Author
-
Alessandrini F, de Jong R, Wimmer M, Maier AM, Fernandez I, Hils M, Buters JT, Biedermann T, Zissler UM, Hoffmann C, Esser-von-Bieren J, Schmidt-Weber CB, and Ohnmacht C
- Subjects
- Allergens, Animals, Cytochrome P-450 Enzyme System, Humans, Inflammation, Mice, Mice, Knockout, Cytochrome P-450 CYP1A1, Lung metabolism, Lung physiopathology, Receptors, Aryl Hydrocarbon genetics, Receptors, Aryl Hydrocarbon metabolism
- Abstract
The lung epithelial barrier serves as a guardian towards environmental insults and responds to allergen encounter with a cascade of immune reactions that can possibly lead to inflammation. Whether the environmental sensor aryl hydrocarbon receptor (AhR) together with its downstream targets cytochrome P450 (CYP1) family members contribute to the regulation of allergic airway inflammation remains unexplored. By employing knockout mice for AhR and for single CYP1 family members, we found that AhR
-/- and CYP1B1-/- but not CYP1A1-/- or CYP1A2-/- animals display enhanced allergic airway inflammation compared to WT. Expression analysis, immunofluorescence staining of murine and human lung sections and bone marrow chimeras suggest an important role of CYP1B1 in non-hematopoietic lung epithelial cells to prevent exacerbation of allergic airway inflammation. Transcriptional analysis of murine and human lung epithelial cells indicates a functional link of AhR to barrier protection/inflammatory mediator signaling upon allergen challenge. In contrast, CYP1B1 deficiency leads to enhanced expression and activity of CYP1A1 in lung epithelial cells and to an increased availability of the AhR ligand kynurenic acid following allergen challenge. Thus, differential CYP1 family member expression and signaling via the AhR in epithelial cells represents an immunoregulatory layer protecting the lung from exacerbation of allergic airway inflammation., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Alessandrini, de Jong, Wimmer, Maier, Fernandez, Hils, Buters, Biedermann, Zissler, Hoffmann, Esser-von-Bieren, Schmidt-Weber and Ohnmacht.)- Published
- 2022
- Full Text
- View/download PDF
258. Macrophages acquire a TNF-dependent inflammatory memory in allergic asthma.
- Author
-
Lechner A, Henkel FDR, Hartung F, Bohnacker S, Alessandrini F, Gubernatorova EO, Drutskaya MS, Angioni C, Schreiber Y, Haimerl P, Ge Y, Thomas D, Kabat AM, Pearce EJ, Ohnmacht C, Nedospasov SA, Murray PJ, Chaker AM, Schmidt-Weber CB, and Esser-von Bieren J
- Subjects
- Animals, Dermatophagoides pteronyssinus, Disease Models, Animal, Humans, Inflammation, Macrophages, Mice, Prostaglandins E metabolism, Pyroglyphidae, Asthma, Hypersensitivity
- Abstract
Background: Infectious agents can reprogram or "train" macrophages and their progenitors to respond more readily to subsequent insults. However, whether such an inflammatory memory exists in type 2 inflammatory conditions such as allergic asthma was not known., Objective: We sought to decipher macrophage-trained immunity in allergic asthma., Methods: We used a combination of clinical sampling of house dust mite (HDM)-allergic patients, HDM-induced allergic airway inflammation in mice, and an in vitro training setup to analyze persistent changes in macrophage eicosanoid, cytokine, and chemokine production as well as the underlying metabolic and epigenetic mechanisms. Transcriptional and metabolic profiles of patient-derived and in vitro trained macrophages were assessed by RNA sequencing or metabolic flux analysis and liquid chromatography-tandem mass spectrometry analysis, respectively., Results: We found that macrophages differentiated from bone marrow or blood monocyte progenitors of HDM-allergic mice or asthma patients show inflammatory transcriptional reprogramming and excessive mediator (TNF-α, CCL17, leukotriene, PGE
2 , IL-6) responses upon stimulation. Macrophages from HDM-allergic mice initially exhibited a type 2 imprint, which shifted toward a classical inflammatory training over time. HDM-induced allergic airway inflammation elicited a metabolically activated macrophage phenotype, producing high amounts of 2-hydroxyglutarate (2-HG). HDM-induced macrophage training in vitro was mediated by a formyl peptide receptor 2-TNF-2-HG-PGE2 /PGE2 receptor 2 axis, resulting in an M2-like macrophage phenotype with high CCL17 production. TNF blockade by etanercept or genetic ablation of Tnf in myeloid cells prevented the inflammatory imprinting of bone marrow-derived macrophages from HDM-allergic mice., Conclusion: Allergen-triggered inflammation drives a TNF-dependent innate memory, which may perpetuate and exacerbate chronic type 2 airway inflammation and thus represents a target for asthma therapy., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
259. Early reduction of SARS-CoV-2-replication in bronchial epithelium by kinin B 2 receptor antagonism.
- Author
-
Jakwerth CA, Feuerherd M, Guerth FM, Oelsner M, Schellhammer L, Giglberger J, Pechtold L, Jerin C, Kugler L, Mogler C, Haller B, Erb A, Wollenberg B, Spinner CD, Buch T, Protzer U, Schmidt-Weber CB, Zissler UM, and Chaker AM
- Subjects
- Animals, Epithelium, Humans, Mice, RNA, Viral, Receptor, Bradykinin B2 genetics, Receptor, Bradykinin B2 metabolism, SARS-CoV-2, COVID-19 Drug Treatment
- Abstract
SARS-CoV-2 has evolved to enter the host via the ACE2 receptor which is part of the kinin-kallikrein pathway. This complex pathway is only poorly understood in context of immune regulation but critical to control infection. This study examines SARS-CoV-2-infection and epithelial mechanisms of the kinin-kallikrein-system at the kinin B
2 receptor level in SARS-CoV-2-infection that is of direct translational relevance. From acute SARS-CoV-2-positive study participants and -negative controls, transcriptomes of nasal curettages were analyzed. Primary airway epithelial cells (NHBEs) were infected with SARS-CoV-2 and treated with the approved B2 R-antagonist icatibant. SARS-CoV-2 RNA RT-qPCR, cytotoxicity assays, plaque assays, and transcriptome analyses were performed. The treatment effect was further studied in a murine airway inflammation model in vivo. Here, we report a broad and strong upregulation of kallikreins and the kinin B2 receptor (B2 R) in the nasal mucosa of acutely symptomatic SARS-CoV-2-positive study participants. A B2 R-antagonist impeded SARS-CoV-2 replication and spread in NHBEs, as determined in plaque assays on Vero-E6 cells. B2 R-antagonism reduced the expression of SARS-CoV-2 entry receptor ACE2, G protein-coupled receptor signaling, and ion transport in vitro and in a murine airway inflammation in vivo model. In summary, this study provides evidence that treatment with B2 R-antagonists protects airway epithelial cells from SARS-CoV-2 by inhibiting its replication and spread, through the reduction of ACE2 levels and the interference with several cellular signaling processes. Future clinical studies need to shed light on the airway protection potential of approved B2 R-antagonists, like icatibant, in the treatment of early-stage COVID-19. KEY MESSAGES: Induction of kinin B2 receptor in the nose of SARS-CoV-2-positive patients. Treatment with B2 R-antagonist protects airway epithelial cells from SARS-CoV-2. B2 R-antagonist reduces ACE2 levels in vivo and ex vivo. Protection by B2 R-antagonist is mediated by inhibiting viral replication and spread., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
260. TGF-β1 Drives Inflammatory Th Cell But Not Treg Cell Compartment Upon Allergen Exposure.
- Author
-
Musiol S, Alessandrini F, Jakwerth CA, Chaker AM, Schneider E, Guerth F, Schnautz B, Grosch J, Ghiordanescu I, Ullmann JT, Kau J, Plaschke M, Haak S, Buch T, Schmidt-Weber CB, and Zissler UM
- Subjects
- Allergens genetics, Animals, Asthma genetics, Asthma pathology, Inflammation genetics, Inflammation immunology, Mice, Mice, Transgenic, Transforming Growth Factor beta1 genetics, Allergens immunology, Asthma immunology, T-Lymphocytes, Helper-Inducer immunology, T-Lymphocytes, Regulatory immunology, Transforming Growth Factor beta1 immunology
- Abstract
TGF-β1 is known to have a pro-inflammatory impact by inducing Th9 and Th17 cells, while it also induces anti-inflammatory Treg cells (Tregs). In the context of allergic airway inflammation (AAI) its dual role can be of critical importance in influencing the outcome of the disease. Here we demonstrate that TGF-β is a major player in AAI by driving effector T cells, while Tregs differentiate independently. Induction of experimental AAI and airway hyperreactivity in a mouse model with inducible genetic ablation of the gene encoding for TGFβ-receptor 2 ( Tgfbr2 ) on CD4
+ T cells significantly reduced the disease phenotype. Further, it blocked the induction of pro-inflammatory T cell frequencies (Th2, Th9, Th17), but increased Treg cells. To translate these findings into a human clinically relevant context, Th2, Th9 and Treg cells were quantified both locally in induced sputum and systemically in blood of allergic rhinitis and asthma patients with or without allergen-specific immunotherapy (AIT). Natural allergen exposure induced local and systemic Th2, Th9, and reduced Tregs cells, while therapeutic allergen exposure by AIT suppressed Th2 and Th9 cell frequencies along with TGF-β and IL-9 secretion. Altogether, these findings support that neutralization of TGF-β represents a viable therapeutic option in allergy and asthma, not posing the risk of immune dysregulation by impacting Tregs cells., Competing Interests: UZ received payment for manuscripts from Deutsches Aerzteblatt and funds for travel from the European Academy of Allergy and Clinical Immunology (EAACI) and Collegium Internationale Allergologicum (CIA). CS-W received support for research projects from PLS Design, LETI, Zeller AG, and Allergopharma and accepted honoraria for consultancy and seminars from LETI and Allergopharma. He also received travel support from EAACI. AC has consultant arrangements through Technical University Munich with Allergopharma, ALK-Abello, HAL Allergy, Mundipharma, and Lofarma; has conducted clinical studies and received research grants through Technical University Munich from Allergopharma, Novartis, the German Federal Environmental Agency, Bencard/Allergen Therapeutics, ASIT Biotech, and Zeller AG; has received payment for lectures from Allergopharma, ALK-Abello, and GlaxoSmithKline; has received payment for manuscript preparation from Bayerisches Ärzteblatt; and has received travel support from the European Academy of Allergy and Clinical Immunology (EAACI), DGAKI, and SMI. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Musiol, Alessandrini, Jakwerth, Chaker, Schneider, Guerth, Schnautz, Grosch, Ghiordanescu, Ullmann, Kau, Plaschke, Haak, Buch, Schmidt-Weber and Zissler.)- Published
- 2022
- Full Text
- View/download PDF
261. Dosing intact birch pollen grains at the air-liquid interface (ALI) to the immortalized human bronchial epithelial cell line BEAS-2B.
- Author
-
Candeias J, Schmidt-Weber CB, and Buters J
- Subjects
- Bronchi chemistry, Bronchi drug effects, Cell Line, Cytokines genetics, Epithelial Cells chemistry, Epithelial Cells cytology, Epithelial Cells drug effects, Fractionation, Field Flow, Gene Expression Regulation, Humans, Interleukin-17 genetics, Interleukin-33 genetics, Pollen adverse effects, Betula chemistry, Bronchi cytology, Gene Expression Profiling methods, Pollen immunology
- Abstract
In real life, humans are exposed to whole pollen grains at the air epithelial barrier. We developed a system for in vitro dosing of whole pollen grains at the Air-Liquid Interface (ALI) and studied their effect on the immortalized human bronchial epithelial cell line BEAS-2B. Pollen are sticky and large particles. Dosing pollen needs resuspension of single particles rather than clusters, and subsequent transportation to the cells with little loss to the walls of the instrumentation i.e. in a straight line. To avoid high speed impacting insults to cells we chose sedimentation by gravity as a delivery step. Pollen was resuspended into single particles by pressured air. A pollen dispersion unit including PTFE coating of the walls and reduced air pressure limited impaction loss to the walls. The loss of pollen to the system was still about 40%. A linear dose effect curve resulted in 327-2834 pollen/cm2 (± 6.1%), the latter concentration being calculated as the amount deposited on epithelial cells on high pollen days. After whole pollen exposure, the largest differential gene expression at the transcriptomic level was late, about 7 hours after exposure. Inflammatory and response to stimulus related genes were up-regulated. We developed a whole pollen exposure air-liquid interface system (Pollen-ALI), in which cells can be gently and reliably dosed., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
262. An exhausted phenotype of T H 2 cells is primed by allergen exposure, but not reinforced by allergen-specific immunotherapy.
- Author
-
Wang SH, Zissler UM, Buettner M, Heine S, Heldner A, Kotz S, Pechtold L, Kau J, Plaschke M, Ullmann JT, Guerth F, Oelsner M, Alessandrini F, Blank S, Chaker AM, Schmidt-Weber CB, and Jakwerth CA
- Subjects
- Allergens, Animals, Cross-Sectional Studies, Humans, Mice, Mice, Inbred C57BL, Phenotype, Prospective Studies, Desensitization, Immunologic, Leukocytes, Mononuclear
- Abstract
Background: Studies show that proallergic T
H 2 cells decrease after successful allergen-specific immunotherapy (AIT). It is likely that iatrogenic administration of allergens drives these cells to exhaustion due to chronic T-cell receptor stimulation. This study aimed to investigate the exhaustion of T cells in connection with allergen exposure during AIT in mice and two independent patient cohorts., Methods: OVA-sensitized C57BL/6J mice were challenged and treated with OVA, and the development of exhaustion in local and systemic TH 2 cells was analyzed. In patients, the expression of exhaustion-associated surface markers on TH 2 cells was evaluated using flow cytometry in a cross-sectional grass pollen allergy cohort with and without AIT. The treatment effect was further studied in PBMC collected from a prospective long-term AIT cohort., Results: The exhaustion-associated surface markers CTLA-4 and PD-1 were significantly upregulated on TH 2 cells upon OVA aerosol exposure in OVA-allergic compared to non-allergic mice. CTLA-4 and PD-1 decreased after AIT, in particular on the surface of local lung TH 2 cells. Similarly, CTLA-4 and PD-1 expression was enhanced on TH 2 cells from patients with allergic rhinitis with an even stronger effect in those with concomitant asthma. Using an unbiased Louvain clustering analysis, we discovered a late-differentiated TH 2 population expressing both markers that decreased during up-dosing but persisted long term during the maintenance phase., Conclusions: This study shows that allergen exposure promotes CTLA-4 and PD-1 expression on TH 2 cells and that the dynamic change in frequencies of exhausted TH 2 cells exhibits a differential pattern during the up-dosing versus the maintenance phases of AIT., (© 2021 Zentrum Allergie und Umwelt. Allergy published by European Academy of Allergy and Clinical Immunology and John Wiley & Sons Ltd.)- Published
- 2021
- Full Text
- View/download PDF
263. Ragweed plants grown under elevated CO 2 levels produce pollen which elicit stronger allergic lung inflammation.
- Author
-
Rauer D, Gilles S, Wimmer M, Frank U, Mueller C, Musiol S, Vafadari B, Aglas L, Ferreira F, Schmitt-Kopplin P, Durner J, Winkler JB, Ernst D, Behrendt H, Schmidt-Weber CB, Traidl-Hoffmann C, and Alessandrini F
- Subjects
- Allergens, Europe, Pollen, Ambrosia, Carbon Dioxide
- Abstract
Background: Common ragweed has been spreading as a neophyte in Europe. Elevated CO
2 levels, a hallmark of global climate change, have been shown to increase ragweed pollen production, but their effects on pollen allergenicity remain to be elucidated., Methods: Ragweed was grown in climate-controlled chambers under normal (380 ppm, control) or elevated (700 ppm, based on RCP4.5 scenario) CO2 levels. Aqueous pollen extracts (RWE) from control- or CO2 -pollen were administered in vivo in a mouse model for allergic disease (daily for 3-11 days, n = 5) and employed in human in vitro systems of nasal epithelial cells (HNECs), monocyte-derived dendritic cells (DCs), and HNEC-DC co-cultures. Additionally, adjuvant factors and metabolites in control- and CO2 -RWE were investigated using ELISA and untargeted metabolomics., Results: In vivo, CO2 -RWE induced stronger allergic lung inflammation compared to control-RWE, as indicated by lung inflammatory cell infiltrate and mediators, mucus hypersecretion, and serum total IgE. In vitro, HNECs stimulated with RWE increased indistinctively the production of pro-inflammatory cytokines (IL-8, IL-1β, and IL-6). In contrast, supernatants from CO2 -RWE-stimulated HNECs, compared to control-RWE-stimulated HNECS, significantly increased TNF and decreased IL-10 production in DCs. Comparable results were obtained by stimulating DCs directly with RWEs. The metabolome analysis revealed differential expression of secondary plant metabolites in control- vs CO2 -RWE. Mixes of these metabolites elicited similar responses in DCs as compared to respective RWEs., Conclusion: Our results indicate that elevated ambient CO2 levels elicit a stronger RWE-induced allergic response in vivo and in vitro and that RWE increased allergenicity depends on the interplay of multiple metabolites., (© 2020 The Authors. Allergy published by European Academy of Allergy and Clinical Immunology and John Wiley & Sons Ltd.)- Published
- 2021
- Full Text
- View/download PDF
264. Inflammatory macrophage memory in nonsteroidal anti-inflammatory drug-exacerbated respiratory disease.
- Author
-
Haimerl P, Bernhardt U, Schindela S, Henkel FDR, Lechner A, Zissler UM, Pastor X, Thomas D, Cecil A, Ge Y, Haid M, Prehn C, Tokarz J, Heinig M, Adamski J, Schmidt-Weber CB, Chaker AM, and Esser-von Bieren J
- Subjects
- Anti-Inflammatory Agents, Non-Steroidal immunology, Asthma chemically induced, Humans, Immunologic Memory immunology, Macrophage Activation immunology, Macrophages metabolism, Nasal Polyps chemically induced, Anti-Inflammatory Agents, Non-Steroidal adverse effects, Asthma immunology, Macrophages immunology, Nasal Polyps immunology
- Abstract
Background: Nonsteroidal anti-inflammatory drug-exacerbated respiratory disease (N-ERD) is a chronic inflammatory condition, which is driven by an aberrant arachidonic acid metabolism. Macrophages are major producers of arachidonic acid metabolites and subject to metabolic reprogramming, but they have been neglected in N-ERD., Objective: This study sought to elucidate a potential metabolic and epigenetic macrophage reprogramming in N-ERD., Methods: Transcriptional, metabolic, and lipid mediator profiles in macrophages from patients with N-ERD and healthy controls were assessed by RNA sequencing, Seahorse assays, and LC-MS/MS. Metabolites in nasal lining fluid, sputum, and plasma from patients with N-ERD (n = 15) and healthy individuals (n = 10) were quantified by targeted metabolomics analyses. Genome-wide methylomics were deployed to define epigenetic mechanisms of macrophage reprogramming in N-ERD., Results: This study shows that N-ERD monocytes/macrophages exhibit an overall reduction in DNA methylation, aberrant metabolic profiles, and an increased expression of chemokines, indicative of a persistent proinflammatory activation. Differentially methylated regions in N-ERD macrophages included genes involved in chemokine signaling and acylcarnitine metabolism. Acylcarnitines were increased in macrophages, sputum, nasal lining fluid, and plasma of patients with N-ERD. On inflammatory challenge, N-ERD macrophages produced increased levels of acylcarnitines, proinflammatory arachidonic acid metabolites, cytokines, and chemokines as compared to healthy macrophages., Conclusions: Together, these findings decipher a proinflammatory metabolic and epigenetic reprogramming of macrophages in N-ERD., (Copyright © 2020 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
265. Near-ground effect of height on pollen exposure.
- Author
-
Rojo J, Oteros J, Pérez-Badia R, Cervigón P, Ferencova Z, Gutiérrez-Bustillo AM, Bergmann KC, Oliver G, Thibaudon M, Albertini R, Rodríguez-De la Cruz D, Sánchez-Reyes E, Sánchez-Sánchez J, Pessi AM, Reiniharju J, Saarto A, Calderón MC, Guerrero C, Berra D, Bonini M, Chiodini E, Fernández-González D, García J, Trigo MM, Myszkowska D, Fernández-Rodríguez S, Tormo-Molina R, Damialis A, Kolek F, Traidl-Hoffmann C, Severova E, Caeiro E, Ribeiro H, Magyar D, Makra L, Udvardy O, Alcázar P, Galán C, Borycka K, Kasprzyk I, Newbigin E, Adams-Groom B, Apangu GP, Frisk CA, Skjøth CA, Radišić P, Šikoparija B, Celenk S, Schmidt-Weber CB, and Buters J
- Subjects
- Allergens, Australia, Europe, Humans, Seasons, Specimen Handling, Environmental Monitoring, Pollen
- Abstract
The effect of height on pollen concentration is not well documented and little is known about the near-ground vertical profile of airborne pollen. This is important as most measuring stations are on roofs, but patient exposure is at ground level. Our study used a big data approach to estimate the near-ground vertical profile of pollen concentrations based on a global study of paired stations located at different heights. We analyzed paired sampling stations located at different heights between 1.5 and 50 m above ground level (AGL). This provided pollen data from 59 Hirst-type volumetric traps from 25 different areas, mainly in Europe, but also covering North America and Australia, resulting in about 2,000,000 daily pollen concentrations analyzed. The daily ratio of the amounts of pollen from different heights per location was used, and the values of the lower station were divided by the higher station. The lower station of paired traps recorded more pollen than the higher trap. However, while the effect of height on pollen concentration was clear, it was also limited (average ratio 1.3, range 0.7-2.2). The standard deviation of the pollen ratio was highly variable when the lower station was located close to the ground level (below 10 m AGL). We show that pollen concentrations measured at >10 m are representative for background near-ground levels., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
266. Type I Immune Response Induces Keratinocyte Necroptosis and Is Associated with Interface Dermatitis.
- Author
-
Lauffer F, Jargosch M, Krause L, Garzorz-Stark N, Franz R, Roenneberg S, Böhner A, Mueller NS, Theis FJ, Schmidt-Weber CB, Biedermann T, Eyerich S, and Eyerich K
- Subjects
- Adolescent, Adult, Aged, Apoptosis immunology, Biopsy, Dermatitis, Atopic genetics, Dermatitis, Atopic pathology, Female, Gene Expression Profiling, Gene Knockdown Techniques, Humans, Keratinocytes immunology, Lichen Planus genetics, Lichen Planus pathology, Lupus Erythematosus, Cutaneous genetics, Lupus Erythematosus, Cutaneous pathology, Male, Middle Aged, Necrosis immunology, Psoriasis genetics, Psoriasis pathology, RNA, Small Interfering metabolism, Receptor-Interacting Protein Serine-Threonine Kinases genetics, Receptor-Interacting Protein Serine-Threonine Kinases immunology, Skin cytology, Skin immunology, Skin pathology, Transcriptome immunology, Dermatitis, Atopic immunology, Keratinocytes pathology, Lichen Planus immunology, Lupus Erythematosus, Cutaneous immunology, Psoriasis immunology
- Abstract
Interface dermatitis is a characteristic histological pattern that occurs in autoimmune and chronic inflammatory skin diseases. It is unknown whether a common mechanism orchestrates this distinct type of skin inflammation. Here we investigated the overlap of two different interface dermatitis positive skin diseases, lichen planus and lupus erythematosus. The shared transcriptome signature pointed toward a strong type I immune response, and biopsy-derived T cells were dominated by IFN-γ and tumor necrosis factor alpha (TNF-α) positive cells. The transcriptome of keratinocytes stimulated with IFN-γ and TNF-α correlated significantly with the shared gene regulations of lichen planus and lupus erythematosus. IFN-γ, TNF-α, or mixed supernatant of lesional T cells induced signs of keratinocyte cell death in three-dimensional skin equivalents. We detected a significantly enhanced epidermal expression of receptor-interacting-protein-kinase 3, a key regulator of necroptosis, in interface dermatitis. Phosphorylation of receptor-interacting-protein-kinase 3 and mixed lineage kinase domain like pseudokinase was induced in keratinocytes on stimulation with T-cell supernatant-an effect that was dependent on the presence of either IFN-γ or TNF-α in the T-cell supernatant. Small hairpin RNA knockdown of receptor-interacting-protein-kinase 3 prevented cell death of keratinocytes on stimulation with IFN-γ or TNF-α. In conclusion, type I immunity is associated with lichen planus and lupus erythematosus and induces keratinocyte necroptosis. These two mechanisms are potentially involved in interface dermatitis., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
267. Toll-like receptor 7/8 agonists stimulate plasmacytoid dendritic cells to initiate T H 17-deviated acute contact dermatitis in human subjects.
- Author
-
Garzorz-Stark N, Lauffer F, Krause L, Thomas J, Atenhan A, Franz R, Roenneberg S, Boehner A, Jargosch M, Batra R, Mueller NS, Haak S, Groß C, Groß O, Traidl-Hoffmann C, Theis FJ, Schmidt-Weber CB, Biedermann T, Eyerich S, and Eyerich K
- Subjects
- Administration, Cutaneous, Adult, Aged, Biomarkers metabolism, Case-Control Studies, Dermatitis, Contact pathology, Female, Flow Cytometry, Humans, Imiquimod administration & dosage, Immunohistochemistry, Male, Middle Aged, Psoriasis pathology, Real-Time Polymerase Chain Reaction, Toll-Like Receptor 8 agonists, Dendritic Cells metabolism, Dermatitis, Contact metabolism, Imiquimod adverse effects, Models, Biological, Psoriasis metabolism, Th17 Cells metabolism, Toll-Like Receptor 7 agonists
- Abstract
Background: A standardized human model to study early pathogenic events in patients with psoriasis is missing. Activation of Toll-like receptor 7/8 by means of topical application of imiquimod is the most commonly used mouse model of psoriasis., Objective: We sought to investigate the potential of a human imiquimod patch test model to resemble human psoriasis., Methods: Imiquimod (Aldara 5% cream; 3M Pharmaceuticals, St Paul, Minn) was applied twice a week to the backs of volunteers (n = 18), and development of skin lesions was monitored over a period of 4 weeks. Consecutive biopsy specimens were taken for whole-genome expression analysis, histology, and T-cell isolation. Plasmacytoid dendritic cells (pDCs) were isolated from whole blood, stimulated with Toll-like receptor 7 agonist, and analyzed by means of extracellular flux analysis and real-time PCR., Results: We demonstrate that imiquimod induces a monomorphic and self-limited inflammatory response in healthy subjects, as well as patients with psoriasis or eczema. The clinical and histologic phenotype, as well as the transcriptome, of imiquimod-induced inflammation in human skin resembles acute contact dermatitis rather than psoriasis. Nevertheless, the imiquimod model mimics the hallmarks of psoriasis. In contrast to classical contact dermatitis, in which myeloid dendritic cells sense haptens, pDCs are primary sensors of imiquimod. They respond with production of proinflammatory and T
H 17-skewing cytokines, resulting in a TH 17 immune response with IL-23 as a key driver. In a proof-of-concept setting systemic treatment with ustekinumab diminished imiquimod-induced inflammation., Conclusion: In human subjects imiquimod induces contact dermatitis with the distinctive feature that pDCs are the primary sensors, leading to an IL-23/TH 17 deviation. Despite these shortcomings, the human imiquimod model might be useful to investigate early pathogenic events and prove molecular concepts in patients with psoriasis., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
268. Virus-like particles (VLP) in prophylaxis and immunotherapy of allergic diseases.
- Author
-
Klimek L, Kündig T, Kramer MF, Guethoff S, Jensen-Jarolim E, Schmidt-Weber CB, Palomares O, Mohsen MO, Jakob T, and Bachmann M
- Abstract
Background: Apart from active allergen avoidance, immunotherapy is regarded as the most effective form of treatment available for type I allergies. Such treatments involve the administration of allergen preparations in various forms and by various routes. Virus-like particles (VLPs) offer a very effective platform for immunization with the allergen and are characterized by high immunogenicity, low allergenicity and high clinical efficacy. Formulations that include Toll-like receptor ligands, T cell stimulatory epitopes and/or depot-forming adjuvants appear to enhance activation of the relevant immune cells. Short nucleotide sequences including CpG motifs have also been intensively explored as potent stimulators of dendritic cells and B cells., Methods: The present paper is based on a systematic literature search in PubMed and MEDLINE, and focuses on the pertinent immunological processes and on clinical data relating to use of VLPs and CpG motifs for the treatment of allergic rhinitis (AR)., Results: Many published studies have reported positive clinical results following administration of VLPs, either alone or in combination with CpG motifs and, in some cases, even in the absence of the allergen-specific allergen., Conclusions: These results indicate that VLPs modulate immune responses in ways which underline their exceptional promise as a platform for the immunotherapy of allergic disorders. However, clinical evaluations remain limited, and further large-scale and longer-term studies will be necessary to substantiate the efficacy and safety of these novel therapies., Competing Interests: L. Klimek has received research support for the execution of studies, and fees for consultancy work, lectures and/or training activities, and/or remuneration as a member of a Scientific Advisory Board from ALK-Abelló, Allergopharma, Bencard, Bionorica, Biomay, Boehringer Ingelheim, Cytos, Circassia, HAL, GSK, Leti, Lofarma, MEDA/Mylan, Novartis, Stallergenes and Roxall. T. Kündig is developing VLPs for clinical use. M.F. Kramer and S. Guethoff are staff members of Bencard Allergie GmbH/Allergy Therapeutics plc; this company is developing VLPs for clinical use. C.B. Schmidt-Weber has received research support, fees for consultancy work, lectures, and travel costs from Allergopharma, Leti Pharma, Bencard, Regeneron, Leo Pharma, Novartis and owns shares of Pls Design and Tolerogenics. T. Jakob has received research support from ALK-Abello, Allergopharma, Cosmetics Europe, Novartis and Thermo Fisher Scientific; and lecturing and consulting fees from ALK-Abello, Allergy Therapeutics/Bencard, Allergopharma, Leti and Novartis. M. Bachmann is developing VLPs for clinical use. E. Jensen-Jarolim, O. Palomares and M.O. Mohsen declare that they have no competing interests.
- Published
- 2018
- Full Text
- View/download PDF
269. Specific Surface Modifications of Silica Nanoparticles Diminish Inflammasome Activation and In Vivo Expression of Selected Inflammatory Genes.
- Author
-
Marzaioli V, Groß CJ, Weichenmeier I, Schmidt-Weber CB, Gutermuth J, Groß O, and Alessandrini F
- Abstract
Silica (SiO₂) nanoparticles (NPs) usage includes, but is not limited to, industrial and biomedical applications. Toxic effects of SiO₂ NPs have been explored either in vitro or in vivo , assessing different surface modifications to reduce their harmful effects. Here, murine bone marrow-derived dendritic (BMDC) and a mouse model of mild allergic inflammation were used to study inflammasome activation and lung inflammation. Our results showed that SiO₂ plain NPs induced NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome activation, increasing interleukin (IL)-1β release in vitro , and, to a lesser extent, in vivo . In addition, SiO₂ plain NPs triggered a pulmonary inflammatory milieu in both non-sensitized (NS) and sensitized (S) mice, by inducing the expression of key inflammatory cytokines and chemokines. Electron microscopy showed that SiO₂ NPs were mostly localized in alveolar macrophages, within vesicles and/or in phagolysosomes. Both the in vitro and the in vivo effects of SiO NPs were attenuated by coating NPs with phosphonate or amino groups, whereas PEGylation, although it mitigated inflammasome activation in vitro , was not a successful coating strategy in vivo . These findings highlight that multiple assays are required to determine the effect of surface modifications in limiting NPs inflammatory potential. Taken together, these data are obtained by comparing in vitro and in vivo effects of SiO₂ NPs suggest the use of amino and phosphonate coating of silica NPs for commercial purposes and targeted applications, as they significantly reduce their proinflammatory potential., Competing Interests: The authors declare no conflict of interest.
- Published
- 2017
- Full Text
- View/download PDF
270. The Use of Adjuvants for Enhancing Allergen Immunotherapy Efficacy.
- Author
-
Chesné J, Schmidt-Weber CB, and Esser von-Bieren J
- Subjects
- Allergens immunology, Animals, Humans, Hypersensitivity immunology, Immune Tolerance, Immunomodulation, Th1-Th2 Balance, Adjuvants, Immunologic therapeutic use, Desensitization, Immunologic, Hypersensitivity therapy
- Abstract
One key approach to increase the efficacy and the safety of immunotherapy is the use of adjuvants. However, many of the adjuvants currently in use can cause adverse events, raising concerns regarding their clinical use, and are geared toward productive immune responses but not necessarily tolerogenic responses. Thus, novel adjuvants for immunotherapy are needed and are being developed. Essential is their potential to boost appropriate tolerogenic adaptive immune responses to allergens while limiting side effects. This review provides an overview of adjuvants currently in clinical use or under development and discusses their therapeutic effect in enhancing allergen-induced tolerance., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
271. A Combined Omics Approach to Generate the Surface Atlas of Human Naive CD4+ T Cells during Early T-Cell Receptor Activation.
- Author
-
Graessel A, Hauck SM, von Toerne C, Kloppmann E, Goldberg T, Koppensteiner H, Schindler M, Knapp B, Krause L, Dietz K, Schmidt-Weber CB, and Suttner K
- Subjects
- Cell Membrane metabolism, Cluster Analysis, Computer Simulation, Flow Cytometry, Gene Expression Profiling, Gene Ontology, Glycoproteins metabolism, Humans, Proteome metabolism, Reproducibility of Results, Transcriptome genetics, CD4-Positive T-Lymphocytes metabolism, Lymphocyte Activation immunology, Proteomics methods, Receptors, Antigen, T-Cell metabolism
- Abstract
Naive CD4(+) T cells are the common precursors of multiple effector and memory T-cell subsets and possess a high plasticity in terms of differentiation potential. This stem-cell-like character is important for cell therapies aiming at regeneration of specific immunity. Cell surface proteins are crucial for recognition and response to signals mediated by other cells or environmental changes. Knowledge of cell surface proteins of human naive CD4(+) T cells and their changes during the early phase of T-cell activation is urgently needed for a guided differentiation of naive T cells and may support the selection of pluripotent cells for cell therapy. Periodate oxidation and aniline-catalyzed oxime ligation technology was applied with subsequent quantitative liquid chromatography-tandem MS to generate a data set describing the surface proteome of primary human naive CD4(+) T cells and to monitor dynamic changes during the early phase of activation. This led to the identification of 173 N-glycosylated surface proteins. To independently confirm the proteomic data set and to analyze the cell surface by an alternative technique a systematic phenotypic expression analysis of surface antigens via flow cytometry was performed. This screening expanded the previous data set, resulting in 229 surface proteins, which were expressed on naive unstimulated and activated CD4(+) T cells. Furthermore, we generated a surface expression atlas based on transcriptome data, experimental annotation, and predicted subcellular localization, and correlated the proteomics result with this transcriptional data set. This extensive surface atlas provides an overall naive CD4(+) T cell surface resource and will enable future studies aiming at a deeper understanding of mechanisms of T-cell biology allowing the identification of novel immune targets usable for the development of therapeutic treatments., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
272. Human IL-31 is induced by IL-4 and promotes TH2-driven inflammation.
- Author
-
Stott B, Lavender P, Lehmann S, Pennino D, Durham S, and Schmidt-Weber CB
- Subjects
- Animals, Bronchi, Cytokines immunology, Cytokines metabolism, Epithelial Cells, Humans, Hypersensitivity, Immediate metabolism, Inflammation metabolism, Interleukin-13 immunology, Interleukin-13 metabolism, Interleukin-4 genetics, Interleukin-4 metabolism, Interleukin-5 immunology, Interleukin-5 metabolism, Interleukins genetics, Interleukins immunology, Lymphocyte Activation immunology, Mice, Gene Expression Regulation, Hypersensitivity, Immediate immunology, Inflammation immunology, Interleukin-4 immunology, Interleukins metabolism, Th2 Cells immunology
- Abstract
Background: The pruritic cytokine IL-31 has been shown to be expressed by murine activated effector T Lymphocytes of a TH2 phenotype. Like IL-17 and IL-22, IL-31 is a tissue-signaling cytokine the receptor of which is mainly found on nonimmune cells. An overabundance of IL-31 has been shown in patients with atopic disorders, including dermatitis, as well as asthma, and therefore represents a promising drug target, although its regulation in the context of the human TH2 clusters is not yet known., Objective: We sought to address the gene regulation of human IL-31 and to test whether IL-31 possesses a similar proallergic function as members of the human TH2 cytokine family, such as IL-4, IL-5, and IL-13., Methods: Polyclonal and purified protein derivative of tuburculin-specific T-cell clones were generated. TH phenotype was determined, and IL-31 was measured by means of ELISA. Gene expression of primary bronchial epithelial cells treated with IL-31 was also measured., Results: IL-31 was expressed by all of the TH2 clones and not by TH1, TH17, or TH22. This expression was dependent on autocrine IL-4 expression from these clones because it could be reduced if blocking antibodies to IL-4 were present. Interestingly, TH1 clones were able to express IL-31 if IL-4 was added to culture. This IL-31 expression was transient and did not affect the phenotype of the TH1 clones. IL-31 was able to induce proinflammatory genes, such as CCL2 and granulocyte colony-stimulating factor., Conclusion: IL-31 is not a TH2 cytokine in the classical sense but is likely to be expressed by a number of cells in an allergic situation in which IL-4 is present and possibly contribute to the allergic reaction., (Copyright © 2013 American Academy of Allergy, Asthma & Immunology. Published by Mosby, Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
273. Barrier responses of human bronchial epithelial cells to grass pollen exposure.
- Author
-
Blume C, Swindle EJ, Dennison P, Jayasekera NP, Dudley S, Monk P, Behrendt H, Schmidt-Weber CB, Holgate ST, Howarth PH, Traidl-Hoffmann C, and Davies DE
- Subjects
- Allergens chemistry, Asthma metabolism, Bronchoscopy, Cells, Cultured, Chemokines immunology, Humans, Inflammation, Interleukin-8 immunology, Ligands, Poaceae, Asthma immunology, Bronchi pathology, Epithelial Cells pathology, Plant Extracts chemistry, Pollen chemistry
- Abstract
The airway epithelium forms a physical, chemical and immunological barrier against inhaled environmental substances. In asthma, these barrier properties are thought to be abnormal. In this study, we analysed the effect of grass pollen on the physical and immunological barrier properties of differentiated human primary bronchial epithelial cells. Following exposure to Timothy grass (Phleum pratense) pollen extract, the integrity of the physical barrier was not impaired as monitored by measuring the transepithelial resistance and immunofluorescence staining of tight junction proteins. In contrast, pollen exposure affected the immunological barrier properties by modulating vectorial mediator release. CXC chemokine ligand (CXCL)8/interleukin (IL)-8 showed the greatest increase in response to pollen exposure with preferential release to the apical compartment. Inhibition of the extracellular signal-regulated kinase 1/2 and p38 mitogen-activated protein kinase pathways selectively blocked apical CXCL8/IL-8 release via a post-transcriptional mechanism. Apical release of CC chemokine ligand (CCL)20/macrophage inflammatory protein-3α, CCL22/monocyte-derived chemokine and tumour necrosis factor-α was significantly increased only in severe asthma cultures, while CCL11/eotaxin-1 and CXCL10/interferon-γ-induced protein-10 were reduced in nonasthmatic cultures. The bronchial epithelial barrier modulates polarised release of mediators in response to pollen without direct effects on its physical barrier properties. The differential response of cells from normal and asthmatic donors suggests the potential for the bronchial epithelium to promote immune dysfunction in asthma.
- Published
- 2013
- Full Text
- View/download PDF
274. IL-22 suppresses IFN-γ-mediated lung inflammation in asthmatic patients.
- Author
-
Pennino D, Bhavsar PK, Effner R, Avitabile S, Venn P, Quaranta M, Marzaioli V, Cifuentes L, Durham SR, Cavani A, Eyerich K, Chung KF, Schmidt-Weber CB, and Eyerich S
- Subjects
- Adult, Asthma metabolism, Bronchi immunology, Bronchi metabolism, Bronchi pathology, Bronchoalveolar Lavage Fluid chemistry, Case-Control Studies, Cell Movement immunology, Cells, Cultured, Chemokine CCL5 immunology, Chemokine CCL5 metabolism, Chemokine CXCL10 immunology, Chemokine CXCL10 metabolism, Epithelial Cells immunology, Epithelial Cells metabolism, Epithelial Cells pathology, Female, Genes, MHC Class I, Genes, MHC Class II, Humans, Intercellular Adhesion Molecule-1 immunology, Intercellular Adhesion Molecule-1 metabolism, Interferon-gamma metabolism, Interleukins metabolism, Male, Pneumonia metabolism, Respiratory Function Tests, T-Lymphocytes metabolism, Wound Healing immunology, Interleukin-22, Asthma immunology, Asthma pathology, Interferon-gamma immunology, Interleukins immunology, Pneumonia immunology, Pneumonia pathology
- Abstract
Background: IL-22 controls tissue homeostasis by both proinflammatory and anti-inflammatory effects. However, the anti-inflammatory mechanisms of IL-22 remain poorly investigated., Objective: We sought to investigate the anti-inflammatory role for IL-22 in human asthma., Methods: T-cell lines derived from lung biopsy specimens of asthmatic patients were characterized by means of flow cytometry. Human bronchial epithelial cells from healthy and asthmatic subjects were stimulated with IL-22, IFN-γ, or the combination of both cytokines. Effects of cytokine stimulation were investigated by using whole-genome analysis, ELISA, and flow cytometry. The functional consequence of cytokine stimulation was evaluated in an in vitro wound repair model and T cell-mediated cytotoxicity experiments. In vivo cytokine expression was measured by using immunohistochemistry and Luminex assays in bronchoalveolar lavage fluid of healthy and asthmatic patients., Results: The current study identifies a tissue-restricted antagonistic interplay of IL-22 and the proinflammatory cytokine IFN-γ. On the one hand, IFN-γ antagonized IL-22-mediated induction of the antimicrobial peptide S100A7 and epithelial cell migration in bronchial epithelial cells. On the other hand, IL-22 decreased epithelial susceptibility to T cell-mediated cytotoxicity by inhibiting the IFN-γ-induced expression of MHC-I, MHC-II, and CD54/intercellular adhesion molecule 1 molecules. Likewise, IL-22 inhibited IFN-γ-induced secretion of the proinflammatory chemokines CCL5/RANTES and CXCL10/interferon-inducible protein 10 in vitro. Consistently, the IL-22 expression in bronchoalveolar lavage fluid of asthmatic patients inversely correlated with the expression of CCL5/RANTES and CXCL10/interferon-inducible protein 10 in vivo., Conclusions: IL-22 might control the extent of IFN-γ-mediated lung inflammation and therefore play a tissue-restricted regulatory role., (Copyright © 2012 American Academy of Allergy, Asthma & Immunology. Published by Mosby, Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
275. IL-22 and TNF-α represent a key cytokine combination for epidermal integrity during infection with Candida albicans.
- Author
-
Eyerich S, Wagener J, Wenzel V, Scarponi C, Pennino D, Albanesi C, Schaller M, Behrendt H, Ring J, Schmidt-Weber CB, Cavani A, Mempel M, Traidl-Hoffmann C, and Eyerich K
- Subjects
- Candida albicans immunology, Chemokines metabolism, Complement C1r metabolism, Complement C1s metabolism, Enzyme-Linked Immunosorbent Assay, Humans, Immunity, Innate, Immunoblotting, Interleukins physiology, Keratinocytes immunology, Male, Mitogen-Activated Protein Kinases metabolism, Polymerase Chain Reaction, S100 Calcium Binding Protein A7, S100 Proteins metabolism, T-Lymphocytes, Helper-Inducer immunology, Transcription Factor AP-1 metabolism, Tumor Necrosis Factor-alpha physiology, beta-Defensins metabolism, Interleukin-22, Candidiasis, Cutaneous immunology, Epidermis immunology, Interleukins immunology, Tumor Necrosis Factor-alpha immunology
- Abstract
T cells exercise their full impact on target cells through a combination of secreted cytokines. The recently described T helper cell subset Th22 is characterized by a combinatorial secretion of IL-22 and TNF-α. Here, we demonstrate that IL-22 increases the TNF-α-dependent induction and secretion of several immune-modulatory molecules such as initial complement factors C1r and C1s, antimicrobial peptides S100A7 and HBD-2 (human β defensin 2), and antimicrobial chemokines CXCL-9/-10/-11 in primary human keratinocytes. The synergism of IL-22 and TNF-α is transmitted intracellularly by MAP kinases and downstream by transcription factors of the AP-1 family. The induction of innate immunity is relevant in an in vitro infection model, where keratinocytes stimulated with Th22 supernatants or recombinant IL-22 plus TNF-α effectively inhibit the growth of Candida albicans and maintain survival of epithelia. Accordingly, the combinatorial stimulation of keratinocytes with IL-22 and TNF-α most efficiently conserves the integrity of the epidermal barrier in a three-dimensional skin infection model as compared with IFN-γ, IL-17, IL-22 or TNF-α alone. In summary, we demonstrate that IL-22 and TNF-α represent a potent, synergistic cytokine combination for cutaneous immunity., (Copyright © 2011 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2011
- Full Text
- View/download PDF
276. Transforming growth factor-beta: recent advances on its role in immune tolerance.
- Author
-
Mantel PY and Schmidt-Weber CB
- Subjects
- Arthritis, Rheumatoid immunology, Base Sequence, CD8 Antigens pharmacology, Cells, Cultured, Diabetes Mellitus, Type 1 immunology, Gene Expression Regulation, Humans, Interleukin-2 pharmacology, Interleukin-4 pharmacology, Malaria immunology, Molecular Sequence Data, Multiple Sclerosis immunology, Promoter Regions, Genetic genetics, Signal Transduction physiology, Transcription Factors genetics, Transforming Growth Factor beta1 immunology, Wound Healing immunology, Cytokines physiology, Forkhead Transcription Factors genetics, Immune Tolerance immunology, Transcription Factors metabolism, Transforming Growth Factor beta physiology
- Abstract
Transforming growth factor (TGF-β1) is a pleiotropic cytokine, secreted by immune and nonhematopoietic cells. TGF-β is involved in many different critical processes, such as embryonal development, cellular maturation and differentiation, wound healing, and immune regulation. It maintains immune homeostasis by acting as a potent immune suppressor through inhibition of proliferation, differentiation, activation, and effector function of immune cells. Paradoxically, depending on the context, it displays proinflammatory properties by being a potent chemoattractant for neutrophils and promoting inflammation. In addition, it does not only induce differentiation into the anti-inflammatory Treg cells, but also into the proinflammatory Th17 and Th9 cells and inhibits Th22 differentiation. TGF-β has been demonstrated to be involved in multiple pathologies. In infections, it protects against collateral damages caused by the immune system, but it also promotes immune evasion and chronic infections. In autoimmune diseases, a TGF-β dysfunction leads to the loss of tolerance to self-antigens. In cancer, TGF-β is a potent inhibitor of cell proliferation and acts as a tumor suppressor at the beginning of tumorogenesis. However, once the cells become resistant to TGF-β, it mainly supports tumor growth and metastasis by promoting immune evasion and angiogenesis. In asthma, it is assumed to promote allergen tolerance, but plays a detrimental role in irreversible remodeling of the airways. Despite the high numbers of TGF-β-targeted pathways, it is a promising drug target for treatment of autoimmunity, cancer, fibrosis, if cell specificity can be achieved.This review summarizes the progresses that have been accomplished on the understanding of TGF-β's signaling in the immune homeostasis and its role in pathogenesis.
- Published
- 2011
- Full Text
- View/download PDF
277. Immunological mechanisms of specific allergen immunotherapy.
- Author
-
Schmidt-Weber CB and Blaser K
- Subjects
- Animals, Humans, Interleukin-10 therapeutic use, Transforming Growth Factor beta physiology, Allergens therapeutic use, Hypersensitivity therapy, Immunotherapy
- Abstract
Allergy is an immunological disorder, which is driven by uncontrolled allergen-activated T cell subsets, leading to immediate type hypersensitivity against otherwise harmless environmental allergens. These allergens are tolerated by healthy individuals as well as by patients, who successfully underwent allergen-specific immunotherapy (SIT). The successful SIT is characterized by the induction of T cell unresponsiveness against the given allergen. Regulatory T cells (Tregs), which are installed or enhanced by SIT and govern the activity of potentially pro-allergic effector T cells, mediate this unresponsiveness. The current article reviews the mechanisms underlying the balance of these cell populations along with suppressive mechanisms of SIT, which may serve as future drug targets.
- Published
- 2006
- Full Text
- View/download PDF
278. TGF-{beta} signaling of human T cells is modulated by the ancillary TGF-{beta} receptor endoglin.
- Author
-
Schmidt-Weber CB, Letarte M, Kunzmann S, Rückert B, Bernabéu C, and Blaser K
- Subjects
- Antigens, CD, Cell Proliferation, Cells, Cultured, Endoglin, Gene Expression Regulation immunology, Humans, Protein Serine-Threonine Kinases, Receptor, Transforming Growth Factor-beta Type I, Receptor, Transforming Growth Factor-beta Type II, Receptors, Cell Surface, Transforming Growth Factor beta immunology, Activin Receptors, Type I immunology, CD4-Positive T-Lymphocytes immunology, Lymphocyte Activation immunology, Receptors, Transforming Growth Factor beta immunology, Signal Transduction immunology, Vascular Cell Adhesion Molecule-1 biosynthesis
- Abstract
Transforming growth factor beta (TGF-beta) inhibits T cell activation and alters differentiation of naive T cells into effector cells. Although four main cell-surface proteins can interact with TGF-beta, only the signaling receptors type I (TGF-betaR type I) and type II (TGF-betaR type II) have so far been described on T cells. The aim of the present study was to investigate the expression of the ancillary receptor endoglin (CD105) by T cells and its role in TGF-beta-mediated signal transduction and function. CD105 expression was analyzed on resting and activated human CD4(+) T cells by flow cytometry, western blot, immunoprecipitation, proliferation and SMAD-responsive reporter gene assays. CD4(+) T cells constitutively expressed CD105 in memory T cells and partially also in naive T cells; however, surface expression is regulated and is increased following TCR engagement, which induced serine/threonine phosphorylation of CD105. In contrast to the suppressive signal mediated by the TGF-beta, cross-linking of CD105 substantially enhanced T cell proliferation, indicating that CD105 by itself mediates signal transduction. Furthermore, CD105 cross-linking induced SMAD-independent signaling via ERK kinase phosphorylation. The present study demonstrates that CD105 is expressed on the surface by activated CD4(+) T cells and CD3 regulated by post-translational means. Furthermore, CD105 acts as a regulatory receptor, counteracting TGF-beta-mediated suppression.
- Published
- 2005
- Full Text
- View/download PDF
279. Expression of cytokine mRNA and protein in joints and lymphoid organs during the course of rat antigen-induced arthritis.
- Author
-
Pohlers D, Siegling A, Buchner E, Schmidt-Weber CB, Palombo-Kinne E, Emmrich F, Bräuer R, and Kinne RW
- Subjects
- Animals, Arthritis, Experimental genetics, Cytokines genetics, Female, Gene Expression Regulation physiology, Joints metabolism, Proteins genetics, RNA, Messenger genetics, Rats, Rats, Inbred Lew, Arthritis, Experimental metabolism, Cytokines biosynthesis, Lymphatic System metabolism, Proteins metabolism, RNA, Messenger biosynthesis, Synovial Membrane metabolism
- Abstract
Cytokine expression was assessed during antigen-induced arthritis (AIA) in synovial membrane (SM), inguinal lymph node (LN), and spleen using competitive RT-PCR and sandwich ELISA. In the SM, early elevations of IL-1beta and IL-6 mRNA (by 6 hours; 450- and 200-fold, respectively) correlated with the joint swelling; a 6-fold increase in tumor necrosis factor alpha (TNFalpha) was not significant. Not only IL-2 and IFN-gamma (which increased 10,000-fold and 200-fold, respectively), but also IL-5 and IL-10, increased acutely (6 hours - day 1; 3-fold and 35-fold, respectively) in the SM. In general, the protein levels in the SM for IL-1beta, IL-6, TNFalpha, IFN-gamma, IL-4, and IL-10 (increase from 4-fold to 15-fold) matched the course of mRNA expression. In the inguinal LN, there were early mRNA elevations of IL-6 (a 2.5-fold increase by 6 hours, which correlated positively with the joint swelling) and IL-2 (4-fold by 6 hours), as well as later rises of IL-4 and IL-5 (2.5- and 4-fold, respectively, by day 3). No significant elevations of the corresponding proteins in this tissue were observed, except for IL-1beta (by day 6) and IL-10 (by day 1). In the spleen, there were significant mRNA elevations at 6 hours of IL-1beta (1.5-fold), IL-6 (4-fold; positively correlated with the joint swelling), IFN-gamma (3-fold), and IL-2 (7- to 10-fold). IL-5 and IL-10 (2- and 3-fold, respectively) peaked from 6 hours to day 3 in the spleen. Increases of the corresponding proteins were significant in comparison with day 0 only in the case of IL-2 (day 6). By day 6 (transition to the chronic phase), the mRNA for cytokines declined to or below prearthritis levels in all the tissues studied except for IL-1beta in the SM and IL-6 in the spleen. AIA is thus characterized by four phenomena: early synovial activation of macrophages, T helper (Th)1-like, and Th2-like cells; late, well-segregated Th2-like responses in the inguinal LN; late, overlapping Th1-like/Th2-like peaks in the spleen; and chronic elevation of synovial IL-1beta mRNA and spleen IL-6 mRNA.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.