Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 290
Filter
Add more filters

Publication year range
1.
Mol Cell ; 77(5): 970-984.e7, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31982308

ABSTRACT

Cytosolic caspase-8 is a mediator of death receptor signaling. While caspase-8 expression is lost in some tumors, it is increased in others, indicating a conditional pro-survival function of caspase-8 in cancer. Here, we show that tumor cells employ DNA-damage-induced nuclear caspase-8 to override the p53-dependent G2/M cell-cycle checkpoint. Caspase-8 is upregulated and localized to the nucleus in multiple human cancers, correlating with treatment resistance and poor clinical outcome. Depletion of caspase-8 causes G2/M arrest, stabilization of p53, and induction of p53-dependent intrinsic apoptosis in tumor cells. In the nucleus, caspase-8 cleaves and inactivates the ubiquitin-specific peptidase 28 (USP28), preventing USP28 from de-ubiquitinating and stabilizing wild-type p53. This results in de facto p53 protein loss, switching cell fate from apoptosis toward mitosis. In summary, our work identifies a non-canonical role of caspase-8 exploited by cancer cells to override the p53-dependent G2/M cell-cycle checkpoint.


Subject(s)
Caspase 8/metabolism , Cell Nucleus/enzymology , Cell Proliferation , G2 Phase Cell Cycle Checkpoints , Neoplasms/enzymology , Tumor Suppressor Protein p53/metabolism , Ubiquitin Thiolesterase/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Caspase 8/genetics , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/pathology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HCT116 Cells , HeLa Cells , Humans , MCF-7 Cells , Male , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , PC-3 Cells , Protein Stability , Signal Transduction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Ubiquitin Thiolesterase/genetics
2.
Immunol Rev ; 314(1): 413-426, 2023 03.
Article in English | MEDLINE | ID: mdl-36331270

ABSTRACT

Neutrophils are the most abundant innate immune cells in human blood, emerging as important players in a variety of diseases. Mitochondria are bioenergetic, biosynthetic, and signaling organelles critical for cell fate and function. Mitochondria have been overlooked in neutrophil research owing to the conventional view that neutrophils contain few, if any, competent mitochondria and do not rely on these organelles for adenosine triphosphate production. A growing body of evidence suggests that mitochondria participate in neutrophil biology at many levels, ranging from neutrophil development to chemotaxis, effector function, and cell death. Moreover, mitochondria and mitochondrial components, such as mitochondrial deoxyribonucleic acid, can be released by neutrophils to eliminate infection and/or shape immune response, depending on the specific context. In this review, we provide an update on the functional role of mitochondria in neutrophils, highlight mitochondria as key players in modulating the neutrophil phenotype and function during infection and inflammation, and discuss the possibilities and challenges to exploit the unique aspects of mitochondria in neutrophils for disease treatment.


Subject(s)
Mitochondria , Neutrophils , Humans , Mitochondria/metabolism , Inflammation , Cell Differentiation , Chemotaxis
3.
Annu Rev Pharmacol Toxicol ; 63: 231-247, 2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36028226

ABSTRACT

Apoptosis is central for the maintenance of health. In the immune system, apoptosis guarantees proper development of immune cells and shutdown of immune reactions by the coordinated elimination of activated immune cells. Limitation of the life span of granulocytes is important, as overactivation of these cells is associated with chronic inflammation and collateral tissue damage. Consequently, targeted induction of granulocyte apoptosis may be beneficial in the course of respective immune disorders. Anti-inflammatory drugs such as glucocorticoids and monoclonal antibodies against IL-5Rα exert their function in part by triggering eosinophil apoptosis. Agonistic antibodies targeting Siglec-8 or death receptors are tested (pre)clinically. Moreover, a new class of inhibitors targeting antiapoptotic BCL-2 proteins shows great promise for anticancer treatments. Because of their specificity and tolerable side effects, these so-called BH3 mimetics may be worthwhile to evaluate in inflammatory disorders. Here, we review past and recent data on pharmacological apoptosis induction of granulocytes and highlight respective therapeutic potential.


Subject(s)
Apoptosis , Eosinophils , Humans , Cell Death , Inflammation/drug therapy
4.
EMBO J ; 40(19): e108863, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34459017

ABSTRACT

Autophagy is a core molecular pathway for the preservation of cellular and organismal homeostasis. Pharmacological and genetic interventions impairing autophagy responses promote or aggravate disease in a plethora of experimental models. Consistently, mutations in autophagy-related processes cause severe human pathologies. Here, we review and discuss preclinical data linking autophagy dysfunction to the pathogenesis of major human disorders including cancer as well as cardiovascular, neurodegenerative, metabolic, pulmonary, renal, infectious, musculoskeletal, and ocular disorders.


Subject(s)
Autophagy , Disease Susceptibility , Animals , Autophagy/drug effects , Autophagy/genetics , Autophagy/immunology , Biomarkers , Gene Expression Regulation , Genetic Predisposition to Disease , Homeostasis , Host-Pathogen Interactions , Humans , Organ Specificity , Signal Transduction
5.
FASEB J ; 38(13): e23730, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38900063

ABSTRACT

Tumor-associated macrophages (TAMs) are integral components of the tumor microenvironment. They are involved in various aspects of tumor cell biology, driving pathological processes such as tumor cell proliferation, metastasis, immunosuppression, and resistance to therapy. TAMs exert their tumorigenic effects by secreting growth factors, cytokines/chemokines, metabolites, and other soluble bioactive molecules. These mediators directly promote tumor cell proliferation and modulate interactions with immune and stromal cells, facilitating further tumor growth. As research into therapies targeting TAMs intensifies, there is a growing need for reliable methods to comprehend the impact of TAMs on cancer progression and to validate novel therapeutics directed at TAMs. The traditional "M1-M2" macrophage classification based on transcriptional profiles of TAMs is not only too simplistic to describe their physiological roles, it also does not explain differences observed between mouse and human macrophages. In this context, methods that assess how TAMs influence tumor or immune cells, either through direct contact or the release of soluble factors, offer a more promising approach. We describe here comprehensive protocols for in vitro functional assays to study TAMs, specifically regarding their impact on the growth of lung cancer cells. We have applied these methods to both mouse and human macrophages, achieving similar outcomes in promoting the proliferation of cancer cells. This methodology can serve as a standardized approach for testing novel therapeutic approaches, targeting TAMs with novel immunotherapeutic compounds, or utilizing gene-editing techniques. Taken together, the described methodology may contribute to our understanding of complex macrophage-tumor interactions and support the development of innovative therapeutic strategies.


Subject(s)
Tumor Microenvironment , Tumor-Associated Macrophages , Humans , Animals , Mice , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Cell Proliferation , Macrophages/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Cell Line, Tumor , Neoplasms/pathology , Neoplasms/metabolism
6.
PLoS Biol ; 20(9): e3001794, 2022 09.
Article in English | MEDLINE | ID: mdl-36108062

ABSTRACT

In contrast to molecular changes associated with increased inflammatory responses, little is known about intracellular counter-regulatory mechanisms that control signaling cascades associated with functional responses of neutrophils. Active RHO GTPases are typically considered as effector proteins that elicit cellular responses. Strikingly, we show here that RHOH, although being constitutively GTP-bound, limits neutrophil degranulation and the formation of neutrophil extracellular traps (NETs). Mechanistically, RHOH is induced under inflammatory conditions and binds to non-muscle myosin heavy chain IIA (NMHC IIA) in activated neutrophils in order to inhibit the transport of mitochondria and granules along actin filaments, which is partially reverted upon disruption of the interaction with NMHC IIA by introducing a mutation in RhoH at lysine 34 (RhoHK34A). In parallel, RHOH inhibits actin polymerization presumably by modulating RAC1 activity. In vivo studies using Rhoh-/- mice, demonstrate an increased antibacterial defense capability against Escherichia coli (E. coli). Collectively, our data reveal a previously undefined role of RHOH as a molecular brake for actomyosin-mediated neutrophil effector functions, which represents an intracellular regulatory axis involved in controlling the strength of an antibacterial inflammatory response.


Subject(s)
Actomyosin , Neutrophils , Transcription Factors , rho GTP-Binding Proteins , Actin Cytoskeleton/metabolism , Actins/metabolism , Actomyosin/metabolism , Animals , Anti-Bacterial Agents , Cytoskeletal Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Guanosine Triphosphate , Lysine , Mice , Myosin Heavy Chains/metabolism , Neutrophils/metabolism , Transcription Factors/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
7.
Allergy ; 79(4): 937-948, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38317432

ABSTRACT

BACKGROUND: Dupilumab is used for the treatment of atopic dermatitis (AD). Approximately one third of AD patients develop a dupilumab-associated ocular surface disease (DAOSD), of which the pathomechanism is poorly understood. This study aimed at investigating inflammatory markers in tear fluids of patients on dupilumab therapy. METHODS: Tear fluids were collected from AD patients with DAOSD (ADwDAOSD), AD patients without DAOSD (ADw/oDAOSD), and non-AD patients before and during dupilumab therapy, and analyzed using a specialized proteomic approach quantifying inflammatory markers. The ocular surface microbiome was determined by next generation sequencing technology. RESULTS: Upon dupilumab therapy, an upregulation of 31 inflammatory markers was observed in DAOSD tear fluids compared to baseline in AD patients. While IL-12B was upregulated in both ADwDAOSD and ADw/oDAOSD groups, the pattern of inflammatory markers significantly differed between groups and over time. In the ADwDAOSD group, a shift from a mixed Th2/Th17 pattern at baseline toward a Th1/Th17 profile under dupilumab was observed. Furthermore, an upregulation of remodeling and fibrosis markers was seen in DAOSD. Semantic map and hierarchical cluster analyses of baseline marker expression revealed four clusters distinguishing between AD and non-AD as well as ADwDAOSD and ADw/oDAOSD patient groups. In a pilot study, dupilumab therapy was associated with a decrease in richness of the ocular surface microbiome. CONCLUSIONS: DAOSD is characterized by a Th1/Th17 cytokine profile and an upregulation of markers known to promote remodeling and fibrosis. The expression pattern of inflammatory markers in tear fluids at baseline might serve as a prognostic factor for DAOSD.


Subject(s)
Antibodies, Monoclonal, Humanized , Dermatitis, Atopic , Eye Diseases , Humans , Pilot Projects , Proteomics , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/metabolism , Inflammation , Fibrosis , Severity of Illness Index , Treatment Outcome
8.
Int Arch Allergy Immunol ; 185(1): 63-72, 2024.
Article in English | MEDLINE | ID: mdl-37866349

ABSTRACT

INTRODUCTION: It is still unknown whether eosinophilic esophagitis (EoE) patients with localized disease are different from those with extended disease. METHODS: We evaluated prospectively included patients in the Swiss EoE cohort. Data on all patients with active disease at baseline, no concomitant gastroesophageal reflux disease, no strictures at baseline, and at least one follow-up visit were analyzed. We compared patients with histologically localized proximal versus distal versus extended (=proximal and distal) disease with regard to patient, disease characteristics, disease presentation, and development of complications. RESULTS: We included 124 patients with a median of 2.5 years of follow-up (73.4% males, median age 35.0 years). Ten patients had proximal (8.1%), 46 patients had distal (37.1%), and 68 patients had extended disease (54.8%). Patients with proximal disease were significantly more often females (80%) compared with patients with distal (26.1%, p = 0.002) or extended disease (19.1%, p < 0.001) and reported less severe symptoms (VAS 0 vs. VAS 1, p = 0.001). Endoscopic and histological disease was less pronounced in the proximal esophagus of proximal EoE compared to extended disease (EREFS 1.0 vs. 3.0, p = 0.001; 27.0 eos/hpf vs. 52.5 eos/hpf, p = 0.008). Patients with proximal disease were less likely to undergo dilation compared to patients with distal disease in the follow-up (3.3% vs. 23.3%, p = 0.010). In a multivariate Cox regression model, proximal eosinophilia was less likely to be associated with treatment failure compared to distal eosinophilia. CONCLUSION: Although isolated proximal EoE is infrequent, it is associated with less severe disease and better disease outcome. Proximal disease appears to present a unique EoE phenotype.


Subject(s)
Enteritis , Eosinophilia , Eosinophilic Esophagitis , Gastritis , Male , Female , Humans , Adult , Eosinophilic Esophagitis/diagnosis , Eosinophilic Esophagitis/epidemiology , Eosinophilic Esophagitis/therapy , Endoscopy , Phenotype
9.
Semin Cancer Biol ; 79: 91-104, 2022 02.
Article in English | MEDLINE | ID: mdl-34280576

ABSTRACT

Beyond their well-known functions in the acute phases of the immune response, neutrophils play important roles in the various phases of tumor initiation and progression, through the release of their stored or newly synthesized mediators. In addition to reactive oxygen species, cytokines, chemokines, granule proteins and lipid mediators, neutrophil extracellular traps (NETs) can also be released upon neutrophil activation. NET formation can be achieved through a cell-death process or in association with the release of mitochondrial DNA from viable neutrophils. NETs are described as extracellular fibers of DNA and decorating proteins responsible for trapping and killing extracellular pathogens, playing a protective role in the antimicrobial defense. There is increasing evidence, however, that NETs play multiple roles in the scenario of cancer-related inflammation. For instance, NETs directly or indirectly promote tumor growth and progression, fostering tumor spread at distant sites and shielding cancer cells thus preventing the effects of cytotoxic lymphocytes. NETs can also promote tumor angiogenesis and cancer-associated thrombosis. On the other hand, there is some evidence that NETs may play anti-inflammatory and anti-tumorigenic roles. In this review, we focus on the main mechanisms underlying the emerging effects of NETs in cancer initiation and progression.


Subject(s)
Carcinogenesis/pathology , Extracellular Traps/immunology , Neoplasms/pathology , Neutrophil Activation/immunology , Neutrophils/immunology , Carcinogenesis/immunology , Cytokines/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Neoplasms/immunology , Neovascularization, Pathologic/pathology
10.
Blood ; 137(21): 2958-2969, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33598715

ABSTRACT

Eosinophils are white blood cells that contribute to the regulation of immunity and are involved in the pathogenesis of numerous inflammatory diseases. In contrast to other cells of the immune system, no information is available regarding the role of autophagy in eosinophil differentiation and functions. To study the autophagic pathway in eosinophils, we generated conditional knockout mice in which Atg5 is deleted within the eosinophil lineage only (designated Atg5eoΔ mice). Eosinophilia was provoked by crossbreeding Atg5eoΔ mice with Il5 (IL-5) overexpressing transgenic mice (designated Atg5eoΔIl5tg mice). Deletion of Atg5 in eosinophils resulted in a dramatic reduction in the number of mature eosinophils in blood and an increase of immature eosinophils in the bone marrow. Atg5-knockout eosinophil precursors exhibited reduced proliferation under both in vitro and in vivo conditions but no increased cell death. Moreover, reduced differentiation of eosinophils in the absence of Atg5 was also observed in mouse and human models of chronic eosinophilic leukemia. Atg5-knockout blood eosinophils exhibited augmented levels of degranulation and bacterial killing in vitro. Moreover, in an experimental in vivo model, we observed that Atg5eoΔ mice achieve better clearance of the local and systemic bacterial infection with Citrobacter rodentium. Evidence for increased degranulation of ATG5low-expressing human eosinophils was also obtained in both tissues and blood. Taken together, mouse and human eosinophil hematopoiesis and effector functions are regulated by ATG5, which controls the amplitude of overall antibacterial eosinophil immune responses.


Subject(s)
Autophagy-Related Protein 5/physiology , Eosinophils/physiology , Myelopoiesis/physiology , Animals , Autophagy-Related Protein 5/biosynthesis , Autophagy-Related Protein 5/deficiency , Autophagy-Related Protein 5/genetics , Bone Marrow/pathology , CRISPR-Cas Systems , Cell Degranulation , Cell Line, Tumor , Cells, Cultured , Citrobacter rodentium , Colony-Forming Units Assay , Enterobacteriaceae Infections/immunology , Eosinophils/cytology , Eosinophils/immunology , Humans , Hypereosinophilic Syndrome/blood , Hypereosinophilic Syndrome/pathology , Interleukin-5/genetics , Leukocyte Count , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Mice, Transgenic , Oncogene Proteins, Fusion/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , mRNA Cleavage and Polyadenylation Factors/genetics
11.
Allergy ; 78(7): 1810-1846, 2023 07.
Article in English | MEDLINE | ID: mdl-37102676

ABSTRACT

Eosinophils are bone marrow-derived granulocytes and are found in low numbers in the peripheral blood of healthy subjects. In type 2 inflammatory diseases, eosinopoiesis in the bone marrow is increased, resulting in a rise in the number of mature eosinophils released in the circulation. From the blood, eosinophils can migrate in multiple tissues and organs under both physiological and pathological conditions. Eosinophils exert their various functions through the synthesis and release of a variety of granule proteins and pro-inflammatory mediators. Despite being present in all species of vertebrates, the functional role of eosinophils is still a matter of debate. Eosinophils may play a role in host defense against various pathogens. In addition, eosinophils have been reported to be involved in tissue homeostasis and exhibit immunomodulatory activities. In this review, we aim to provide a broad overview of eosinophil biology and eosinophilic diseases in a lexicon-style format using keywords starting from A until Z with cross-references to other chapters indicated in italics in the text or specified in parentheses.


Subject(s)
Eosinophils , Animals , Humans , Eosinophils/physiology , Italy
12.
Allergy ; 78(1): 47-59, 2023 01.
Article in English | MEDLINE | ID: mdl-36207764

ABSTRACT

Eosinophilia and eosinophil activation are recurrent features in various reactive states and certain hematologic malignancies. In patients with hypereosinophilia (HE), HE-induced organ damage is often encountered and may lead to the diagnosis of a hypereosinophilic syndrome (HES). A number of known mechanisms and etiologies contribute to the development of HE and HES. Based on these etiologies and the origin of eosinophils, HE and HES are divided into primary forms where eosinophils are clonal cells, reactive forms where an underlying reactive or neoplastic condition is detected and eosinophils are considered to be "non-clonal" cells, and idiopathic HE and HES in which neither a clonal nor a reactive underlying pathology is detected. Since 2012, this classification and the related criteria have been widely accepted and regarded as standard. However, during the past few years, new developments in the field and an increasing number of markers and targets have created a need to update these criteria and the classification of HE and HES. To address this challenge, a Working Conference on eosinophil disorders was organized in 2021. In this conference, a panel of experts representing the relevant fields, including allergy, dermatology, hematology, immunology, laboratory medicine, and pathology, met and discussed new markers and concepts as well as refinements in definitions, criteria and classifications of HE and HES. The outcomes of this conference are presented in this article and should assist in the diagnosis and management of patients with HE and HES in daily practice and in the preparation and conduct of clinical trials.


Subject(s)
Eosinophilia , Hypereosinophilic Syndrome , Hypersensitivity , Humans , Eosinophils/pathology , Eosinophilia/diagnosis , Eosinophilia/etiology , Eosinophilia/drug therapy , Syndrome , Hypersensitivity/complications , Hypereosinophilic Syndrome/etiology , Hypereosinophilic Syndrome/complications
13.
Allergy ; 78(12): 3077-3102, 2023 12.
Article in English | MEDLINE | ID: mdl-37702095

ABSTRACT

Over the past years, eosinophils have become a focus of scientific interest, especially in the context of their recently uncovered functions (e.g. antiviral, anti-inflammatory, regulatory). These versatile cells display both beneficial and detrimental activities under various physiological and pathological conditions. Eosinophils are involved in the pathogenesis of many diseases which can be classified into primary (clonal) and secondary (reactive) disorders and idiopathic (hyper)eosinophilic syndromes. Depending on the biological specimen, the eosinophil count in different body compartments may serve as a biomarker reflecting the underlying pathophysiology and/or activity of distinct diseases and as a therapy-driving (predictive) and monitoring tool. Personalized selection of an appropriate therapeutic strategy directly or indirectly targeting the increased number and/or activity of eosinophils should be based on the understanding of eosinophil homeostasis including their interactions with other immune and non-immune cells within different body compartments. Hence, restoring as well as maintaining homeostasis within an individual's eosinophil pool is a goal of both specific and non-specific eosinophil-targeting therapies. Despite the overall favourable safety profile of the currently available anti-eosinophil biologics, the effect of eosinophil depletion should be monitored from the perspective of possible unwanted consequences.


Subject(s)
Eosinophils , Humans , Biomarkers
14.
Mol Cell ; 57(6): 1011-1021, 2015 Mar 19.
Article in English | MEDLINE | ID: mdl-25728769

ABSTRACT

Eosinophils are white blood cells that function in innate immunity and participate in the pathogenesis of various inflammatory and neoplastic disorders. Their secretory granules contain four cytotoxic proteins, including the eosinophil major basic protein (MBP-1). How MBP-1 toxicity is controlled within the eosinophil itself and activated upon extracellular release is unknown. Here we show how intragranular MBP-1 nanocrystals restrain toxicity, enabling its safe storage, and characterize them with an X-ray-free electron laser. Following eosinophil activation, MBP-1 toxicity is triggered by granule acidification, followed by extracellular aggregation, which mediates the damage to pathogens and host cells. Larger non-toxic amyloid plaques are also present in tissues of eosinophilic patients in a feedback mechanism that likely limits tissue damage under pathological conditions of MBP-1 oversecretion. Our results suggest that MBP-1 aggregation is important for innate immunity and immunopathology mediated by eosinophils and clarify how its polymorphic self-association pathways regulate toxicity intra- and extracellularly.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Eosinophils/metabolism , Animals , Cell Death/drug effects , Cell Line/drug effects , Cell Membrane/drug effects , Cellulitis/metabolism , Cellulitis/pathology , DNA-Binding Proteins/toxicity , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/pathology , Eosinophilia/metabolism , Eosinophilia/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Escherichia coli/drug effects , Host-Pathogen Interactions , Humans , Immunity, Innate/physiology , Mice, Inbred C57BL , Nanoparticles/metabolism , Nanoparticles/toxicity , Secretory Vesicles/metabolism , Skin/drug effects , Skin/pathology
15.
J Allergy Clin Immunol ; 150(2): 251-258, 2022 08.
Article in English | MEDLINE | ID: mdl-35934679

ABSTRACT

With the development of targeted therapies for allergic diseases, the need for biomarkers supporting disease diagnosis and management has increased. Recent research has elucidated the pattern of cytokines and their distinct roles in the pathogenesis of allergic diseases. This means that cytokines should be considered as biomarkers. In this review article, we summarize published findings and critically discuss the use of cytokine measurements in association with disease diagnosis and management. Among the variety of suggested cytokines, thymus and activation-regulated chemokine (TARC) stands out and can indeed serve as a biomarker of atopic dermatitis. Both biologic characteristics and technical issues determine the reliability and limit the use of blood cytokines as biomarkers.


Subject(s)
Chemokine CCL17 , Cytokines , Biomarkers , Humans , Reproducibility of Results , Severity of Illness Index
16.
Int J Mol Sci ; 24(3)2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36768969

ABSTRACT

Neutrophil extracellular traps (NETs) and oxidative stress are considered to be beneficial in the innate immune defense against pathogens. However, defective clearance of NETs in the lung of acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected patients could lead to severe respiratory syndrome infection, the so-called coronavirus disease 2019 (COVID-19). To elucidate the pathways that are related to NETs within the pathophysiology of COVID-19, we utilized RNA sequencing (RNA-seq) as well as immunofluorescence and immunohistochemistry methods. RNA-seq analysis provided evidence for increased oxidative stress and the activation of viral-related signaling pathways in post-mortem lungs of COVID-19 patients compared to control donors. Moreover, an excess of neutrophil infiltration and NET formation were detected in the patients' lungs, where the extracellular DNA was oxidized and co-localized with neutrophil granule protein myeloperoxidase (MPO). Interestingly, staining of the lipid peroxidation marker 4-hydroxynonenal (4-HNE) depicted high colocalization with NETs and was correlated with the neutrophil infiltration of the lung tissues, suggesting that it could serve as a suitable marker for the identification of NETs and the severity of the disease. Moreover, local inhalation therapy to reduce the excess lipid oxidation and NETs in the lungs of severely infected patients might be useful to ameliorate their clinical conditions.


Subject(s)
COVID-19 , Extracellular Traps , Humans , COVID-19/metabolism , Extracellular Traps/metabolism , SARS-CoV-2 , Lung , Oxidative Stress , Neutrophils/metabolism
17.
Allergy ; 77(8): 2520-2533, 2022 08.
Article in English | MEDLINE | ID: mdl-35094416

ABSTRACT

OBJECTIVE: Physicians are increasingly confronted with patients presenting with symptoms of esophageal dysfunction resembling eosinophilic esophagitis (EoE), but absence of significant esophageal eosinophilia. The purpose of this study was to characterize and classify this group of EoE variants. DESIGN: Patients from six EoE-centers with symptoms of esophageal dysfunction, but peak eosinophil counts of <60/mm2 (<15/hpf) in esophageal biopsies and absence of gastro-esophageal reflux disease (GERD) were included. Clinical, endoscopic, (immuno)-histological, and molecular features were determined and compared with EoE, GERD, and healthy controls. RESULTS: We included 69 patients with EoE variants. Endoscopic abnormalities were found in 53.6%. We identified three histological subtypes: EoE-like esophagitis (36/69, 52.2%), lymphocytic esophagitis (14/69, 20.3%), and non-specific esophagitis (19/69, 27.5%). Immunohistochemistry revealed-in contrast to EoE-no significant increase in inflammatory cell infiltrates compared with GERD and healthy controls, except for lymphocytes in lymphocytic esophagitis. EoE-typical Th2-response was absent in all EoE variants. However, considerable structural changes were detected based on histology and protein expression. Using next generation mRNA sequencing, we found the three EoE variants to have distinct molecular fingerprints partially sharing pronounced traits of EoE. Hierarchical sample clustering of RNA sequencing data confirmed the presence of an EoE-like (characterized by eotaxin-3 expression), non-specific, and lymphocytic variant cluster (characterized by CD3 cells and TSLP expression). CONCLUSION: All EoE variants are clinically and histologically active conditions despite the absence of esophageal eosinophilia. EoE variants appear to be part of a disease spectrum, where classical EoE represents the most common and apparent phenotype.


Subject(s)
Eosinophilic Esophagitis , Gastroesophageal Reflux , Cross-Sectional Studies , Enteritis , Eosinophilia , Eosinophilic Esophagitis/diagnosis , Eosinophilic Esophagitis/genetics , Eosinophilic Esophagitis/metabolism , Eosinophils/metabolism , Gastritis , Gastroesophageal Reflux/diagnosis , Gastroesophageal Reflux/genetics , Gastroesophageal Reflux/pathology , Humans
18.
J Immunol ; 205(6): 1653-1663, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32796025

ABSTRACT

Cytokine-primed neutrophils can undergo a nonapoptotic type of cell death using components of the necroptotic pathway, including receptor-interacting protein kinase-3 (RIPK3), mixed lineage kinase-like (MLKL) and NADPH oxidase. In this report, we provide evidence for a potential role of serine proteases in CD44-mediated necroptotic death of GM-CSF-primed human neutrophils. Specifically, we observed that several inhibitors known to block the enzymatic function of fibroblast activation protein-α (FAP-α) were able to block CD44-mediated reactive oxygen species production and cell death, but not FAS receptor-mediated apoptosis. To understand how FAP-α is involved in this nonapoptotic death pathway, we performed immunoblotting experiments in the presence and absence of inhibitors of RIPK3, MLKL, p38 MAPK, PI3K, and FAP-α. The results of these experiments suggested that FAP-α is active in parallel with RIPK3, MLKL, and p38 MAPK activation but proximal to PI3K and NADPH oxidase activation. Interestingly, neutrophils isolated from the joints of patients suffering from rheumatoid arthritis underwent a GM-CSF-independent necroptosis following CD44 ligation; this effect was also blocked by both FAP-α and MLKL inhibitors. Taken together, our evidence shows that the RIPK3-MLKL pathway activates NADPH oxidase but requires, in addition to p38 MAPK and PI3K, a serine protease activity, whereby FAP-α is the most likely candidate. Thus, FAP-α could be a potential drug target in neutrophilic inflammatory responses to avoid exaggerated nonapoptotic neutrophil death, leading to tissue damage.


Subject(s)
Arthritis, Rheumatoid/immunology , Gelatinases/metabolism , Membrane Proteins/metabolism , Neutrophils/metabolism , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Serine Endopeptidases/metabolism , Cells, Cultured , Endopeptidases , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Molecular Targeted Therapy , NADPH Oxidases/metabolism , Necroptosis , Neutrophil Activation , Neutrophils/immunology , Signal Transduction , fas Receptor/metabolism
19.
J Immunol ; 205(10): 2640-2648, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33008951

ABSTRACT

IVIG preparations consisting of pooled IgG are increasingly used for the treatment of autoimmune diseases. IVIG is known to regulate the viability of immune cells, including neutrophils. We report that plasma-derived IgA efficiently triggers death of neutrophils primed by cytokines or TLR agonists. IgA-mediated programmed neutrophil death was PI3K-, p38 MAPK-, and JNK-dependent and evoked anti-inflammatory cytokines in macrophage cocultures. Neutrophils from patients with acute Crohn's disease, rheumatoid arthritis, or sepsis were susceptible to both IgA- and IVIG-mediated death. In contrast to IVIG, IgA did not promote cell death of quiescent neutrophils. Our findings suggest that plasma-derived IgA might provide a therapeutic option for the treatment of neutrophil-associated inflammatory disorders.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Crohn Disease/drug therapy , Immunoglobulin A/pharmacology , Neutrophils/drug effects , Sepsis/drug therapy , Animals , Apoptosis/drug effects , Apoptosis/immunology , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Coculture Techniques , Crohn Disease/blood , Crohn Disease/immunology , Humans , Immunoglobulin A/therapeutic use , Immunoglobulins, Intravenous/pharmacology , Immunoglobulins, Intravenous/therapeutic use , Macrophages , Mice , Neutrophils/immunology , Primary Cell Culture , Sepsis/blood , Sepsis/immunology
20.
J Allergy Clin Immunol ; 148(2): 327-335, 2021 08.
Article in English | MEDLINE | ID: mdl-33895002

ABSTRACT

Chronic rhinosinusitis (CRS) often is characterized by an eosinophilic inflammatory pattern, nowadays referred to as type 2 inflammation, although the mucosal inflammation is dominated by neutrophils in about a third of the patients. Neutrophils are typically predominant in 50% of patients with CRS without nasal polyps, but also are found to play a role in patients with severe type 2 CRS with nasal polyp disease. This review aims at summarizing the current understanding of the eosinophilic and neutrophilic inflammation in CRS pathophysiology, and provides a discussion of their reciprocal interactions and the clinical impact of the mixed presentation in patients with severe type 2 CRS with nasal polyps. A solid understanding of these interactions is of utmost importance when treating uncontrolled severe CRS with nasal polyps with biologicals that are preferentially directed toward type 2 inflammation. We here focus on recent findings on both eosinophilic and neutrophilic granulocytes, their subgroups and the activation status, and their interactions in CRS.


Subject(s)
Eosinophils/immunology , Neutrophils/immunology , Rhinitis/immunology , Sinusitis/immunology , Chronic Disease , Eosinophils/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Inflammation/therapy , Nasal Polyps/immunology , Nasal Polyps/pathology , Nasal Polyps/therapy , Neutrophils/pathology , Rhinitis/pathology , Rhinitis/therapy , Severity of Illness Index , Sinusitis/pathology , Sinusitis/therapy
SELECTION OF CITATIONS
SEARCH DETAIL