Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Paul Peeters is active.

Publication


Featured researches published by Paul Peeters.


Particle and Fibre Toxicology | 2013

Silica induces NLRP3 inflammasome activation in human lung epithelial cells

Paul Peeters; Timothy N Perkins; Emiel F.M. Wouters; Brooke T. Mossman; Niki L. Reynaert

BackgroundIn myeloid cells the inflammasome plays a crucial role in innate immune defenses against pathogen- and danger-associated patterns such as crystalline silica. Respirable mineral particles impinge upon the lung epithelium causing irreversible damage, sustained inflammation and silicosis. In this study we investigated lung epithelial cells as a target for silica-induced inflammasome activation.MethodsA human bronchial epithelial cell line (BEAS-2B) and primary normal human bronchial epithelial cells (NHBE) were exposed to toxic but nonlethal doses of crystalline silica over time to perform functional characterization of NLRP3, caspase-1, IL-1β, bFGF and HMGB1. Quantitative RT-PCR, caspase-1 enzyme activity assay, Western blot techniques, cytokine-specific ELISA and fibroblast (MRC-5 cells) proliferation assays were performed.ResultsWe were able to show transcriptional and translational upregulation of the components of the NLRP3 intracellular platform, as well as activation of caspase-1. NLRP3 activation led to maturation of pro-IL-1β to secreted IL-1β, and a significant increase in the unconventional release of the alarmins bFGF and HMGB1. Moreover, release of bFGF and HMGB1 was shown to be dependent on particle uptake. Small interfering RNA experiments using siNLRP3 revealed the pivotal role of the inflammasome in diminished release of pro-inflammatory cytokines, danger molecules and growth factors, and fibroblast proliferation.ConclusionOur novel data indicate the presence and functional activation of the NLRP3 inflammasome by crystalline silica in human lung epithelial cells, which prolongs an inflammatory signal and affects fibroblast proliferation, mediating a cadre of lung diseases.


Particle and Fibre Toxicology | 2012

Differences in gene expression and cytokine production by crystalline vs. amorphous silica in human lung epithelial cells

Timothy N Perkins; Arti Shukla; Paul Peeters; Jeremy L. Steinbacher; Christopher C. Landry; Sherrill A. Lathrop; Chad Steele; Niki L. Reynaert; Emiel F.M. Wouters; Brooke T. Mossman

BackgroundExposure to respirable crystalline silica particles, as opposed to amorphous silica, is associated with lung inflammation, pulmonary fibrosis (silicosis), and potentially with lung cancer. We used Affymetrix/GeneSifter microarray analysis to determine whether gene expression profiles differed in a human bronchial epithelial cell line (BEAS 2B) exposed to cristobalite vs. amorphous silica particles at non-toxic and equal surface areas (75 and 150 × 106μm2/cm2). Bio-Plex analysis was also used to determine profiles of secreted cytokines and chemokines in response to both particles. Finally, primary human bronchial epithelial cells (NHBE) were used to comparatively assess silica particle-induced alterations in gene expression.ResultsMicroarray analysis at 24 hours in BEAS 2B revealed 333 and 631 significant alterations in gene expression induced by cristobalite at low (75) and high (150 × 106μm2/cm2) amounts, respectively (p < 0.05/cut off ≥ 2.0-fold change). Exposure to amorphous silica micro-particles at high amounts (150 × 106μm2/cm2) induced 108 significant gene changes. Bio-Plex analysis of 27 human cytokines and chemokines revealed 9 secreted mediators (p < 0.05) induced by crystalline silica, but none were induced by amorphous silica. QRT-PCR revealed that cristobalite selectively up-regulated stress-related genes and cytokines (FOS, ATF3, IL6 and IL8) early and over time (2, 4, 8, and 24 h). Patterns of gene expression in NHBE cells were similar overall to BEAS 2B cells. At 75 × 106μm2/cm2, there were 339 significant alterations in gene expression induced by cristobalite and 42 by amorphous silica. Comparison of genes in response to cristobalite (75 × 106μm2/cm2) revealed 60 common, significant gene alterations in NHBE and BEAS 2B cells.ConclusionsCristobalite silica, as compared to synthetic amorphous silica particles at equal surface area concentrations, had comparable effects on the viability of human bronchial epithelial cells. However, effects on gene expression, as well as secretion of cytokines and chemokines, drastically differed, as the crystalline silica induced more intense responses. Our studies indicate that toxicological testing of particulates by surveying viability and/or metabolic activity is insufficient to predict their pathogenicity. Moreover, they show that acute responses of the lung epithelium, including up-regulation of genes linked to inflammation, oxidative stress, and proliferation, as well as secretion of inflammatory and proliferative mediators, can be indicative of pathologic potential using either immortalized lines (BEAS 2B) or primary cells (NHBE). Assessment of the degree and magnitude of these responses in vitro are suggested as predictive in determining the pathogenicity of potentially harmful particulates.


Particle and Fibre Toxicology | 2014

Silica-induced NLRP3 inflammasome activation in vitro and in rat lungs

Paul Peeters; Irene M. J. Eurlings; Timothy N Perkins; Emiel F.M. Wouters; Roel P. F. Schins; Paul J. A. Borm; Wolfgang Drommer; Niki L. Reynaert; Catrin Albrecht

RationaleMineral particles in the lung cause inflammation and silicosis. In myeloid and bronchial epithelial cells the inflammasome plays a role in responses to crystalline silica. Thioredoxin (TRX) and its inhibitory protein TRX-interacting protein link oxidative stress with inflammasome activation. We investigated inflammasome activation by crystalline silica polymorphs and modulation by TRX in vitro, as well as its localization and the importance of silica surface reactivity in rats.MethodsWe exposed bronchial epithelial cells and differentiated macrophages to silica polymorphs quartz and cristobalite and measured caspase-1 activity as well as the release of IL-1β, bFGF and HMGB1; including after TRX overexpression or treatment with recombinant TRX. Rats were intratracheally instilled with vehicle control, Dörentruper quartz (DQ12) or DQ12 coated with polyvinylpyridine N-oxide. At days 3, 7, 28, 90, 180 and 360 five animals per treatment group were sacrificed. Hallmarks of silicosis were assessed with Haematoxylin-eosin and Sirius Red stainings. Caspase-1 activity in the bronchoalveolar lavage and caspase-1 and IL-1β localization in lung tissue were determined using Western blot and immunohistochemistry (IHC).ResultsSilica polymorphs triggered secretion of IL-1β, bFGF and HMGB1 in a surface reactivity dependent manner. Inflammasome readouts linked with caspase-1 enzymatic activity were attenuated by TRX overexpression or treatment. At day 3 and 7 increased caspase-1 activity was detected in BALF of the DQ12 group and increased levels of caspase-1 and IL-1β were observed with IHC in the DQ12 group compared to controls. DQ12 exposure revealed silicotic nodules at 180 and 360 days. Particle surface modification markedly attenuated the grade of inflammation and lymphocyte influx and attenuated the level of inflammasome activation, indicating that the development of silicosis and inflammasome activation is determined by crystalline silica surface reactivity.ConclusionOur novel data indicate the pivotal role of surface reactivity of crystalline silica to activate the inflammasome in cultures of both epithelial cells and macrophages. Inhibitory capacity of the antioxidant TRX to inflammasome activation was evidenced. DQ12 quartz exposure induced acute and chronic functional activation of the inflammasome in the heterogeneous cell populations of the lung in associated with its crystalline surface reactivity.


Inflammatory Bowel Diseases | 2015

Strong Upregulation of AIM2 and IFI16 Inflammasomes in the Mucosa of Patients with Active Inflammatory Bowel Disease

Wiebe Vanhove; Paul Peeters; Dominiek Staelens; Anica Schraenen; Jan Van der Goten; Isabelle Cleynen; Sebastiaan De Schepper; Leentje Van Lommel; Niki L. Reynaert; Frans Schuit; Gert Van Assche; Marc Ferrante; Gert De Hertogh; Emiel F.M. Wouters; Paul Rutgeerts; Severine Vermeire; Kris Nys; Ingrid Arijs

Background:Inflammatory bowel disease (IBD) is characterized by a chronic inflammation of the gut, partly driven by defects in the innate immune system. Considering the central role of inflammasome signaling in innate immunity, we studied inflammasome components in IBD mucosa. Methods:Expression of genes encoding inflammasome sensor subunits was investigated in colonic mucosal biopsies from 2 cohorts of patients with IBD and controls. Results:A significant upregulation (>2-fold change in expression, false discovery rate <0.05) of the PYHIN inflammasomes AIM2 and IFI16 in active IBD versus controls was found. Also IFI16 was significantly increased in inactive IBD versus controls. Moreover, responders to anti-tumor necrosis factor therapy showed decreased expression of these inflammasomes although IFI16 remained significantly increased in responders showing endoscopic healing versus controls. AIM2 was mainly expressed in epithelial cells, whereas IFI16 was expressed in both lymphocytes and epithelial cells. Functional activation of predominant AIM2/IFI16-mediated inflammasomes in active IBD colon was shown by the presence of the downstream effectors CASP1 and HMGB-1 in inflamed mucosa. Conclusions:Our results highlight the importance of PYHIN inflammasome signaling in IBD and also link anti-tumor necrosis factor responsiveness to inflammasome signaling. Together, this points to the potential value of the inflammasome pathway as a new therapeutic target for IBD treatment.


Human Molecular Genetics | 2015

Indications for distinct pathogenic mechanisms of asbestos and silica through gene expression profiling of the response of lung epithelial cells

Timothy N Perkins; Paul Peeters; Arti Shukla; Ingrid Arijs; Julie Dragon; Emiel F.M. Wouters; Niki L. Reynaert; Brooke T. Mossman

Occupational and environmental exposures to airborne asbestos and silica are associated with the development of lung fibrosis in the forms of asbestosis and silicosis, respectively. However, both diseases display distinct pathologic presentations, likely associated with differences in gene expression induced by different mineral structures, composition and bio-persistent properties. We hypothesized that effects of mineral exposure in the airway epithelium may dictate deviating molecular events that may explain the different pathologies of asbestosis versus silicosis. Using robust gene expression-profiling in conjunction with in-depth pathway analysis, we assessed early (24 h) alterations in gene expression associated with crocidolite asbestos or cristobalite silica exposures in primary human bronchial epithelial cells (NHBEs). Observations were confirmed in an immortalized line (BEAS-2B) by QRT-PCR and protein assays. Utilization of overall gene expression, unsupervised hierarchical cluster analysis and integrated pathway analysis revealed gene alterations that were common to both minerals or unique to either mineral. Our findings reveal that both minerals had potent effects on genes governing cell adhesion/migration, inflammation, and cellular stress, key features of fibrosis. Asbestos exposure was most specifically associated with aberrant cell proliferation and carcinogenesis, whereas silica exposure was highly associated with additional inflammatory responses, as well as pattern recognition, and fibrogenesis. These findings illustrate the use of gene-profiling as a means to determine early molecular events that may dictate pathological processes induced by exogenous cellular insults. In addition, it is a useful approach for predicting the pathogenicity of potentially harmful materials.


Clinical & Developmental Immunology | 2015

Immune Homeostasis in Epithelial Cells: Evidence and Role of Inflammasome Signaling Reviewed

Paul Peeters; Emiel F.M. Wouters; Niki L. Reynaert

The epithelium regulates the interaction between the noxious xenogenous, as well as the microbial environment and the immune system, not only by providing a barrier but also by expressing a number of immunoregulatory membrane receptors, and intracellular danger sensors and their downstream effectors. Amongst these are a number of inflammasome sensor subtypes, which have been initially characterized in myeloid cells and described to be activated upon assembly into multiprotein complexes by microbial and environmental triggers. This review compiles a vast amount of literature that supports a pivotal role for inflammasomes in the various epithelial barriers of the human body as essential factors maintaining immune signaling and homeostasis.


Reference Module in Biomedical Sciences#R##N#Pathobiology of Human Disease#R##N#A Dynamic Encyclopedia of Disease Mechanisms | 2014

Pathogenesis and Mechanisms of Asbestosis and Silicosis

T.N. Perkins; Paul Peeters; E.F.M. Wouters; N.L. Reynaert; B.T. Mossman

1. Asbestosis and silicosis are occupational diseases of the lung interstitium characterized by an initial phase of epithelial injury and hyperplasia, followed by an accumulation of cells of the immune system, and the development of fibrosis. Once established, these diseases can progress in the absence of further exposures to asbestos fibers or silica particles. These particulates exist in different physicochemical forms that may be modified by interactions with other minerals or elements such as iron in the lung.


Toxicology and Applied Pharmacology | 2018

Crystalline silica alters Sulfatase-1 expression in rat lungs which influences hyper-proliferative and fibrogenic effects in human lung epithelial cells

Timothy N. Perkins; Paul Peeters; Catrin Albrecht; Roel P. F. Schins; Mieke A. Dentener; Brooke T. Mossman; Emiel F.M. Wouters; Niki L. Reynaert

ABSTRACT Lung epithelial cells are the first cell‐type to come in contact with hazardous dust materials. Upon deposition, they invoke complex reactions in attempt to eradicate particles from the airways, and repair damage. The cell surface is composed of a heterogeneous network of matrix proteins and proteoglycans, which act as scaffold and control cell‐signaling networks. These functions are controlled, in part, by the sulfation patterns of heparin‐sulfate proteoglycans (HSPGs), which are enzymatically regulated. Although there is evidence of altered HSPG‐sulfation in idiopathic pulmonary fibrosis (IPF), this is not investigated in silicosis. Our previous studies revealed down‐regulation of Sulfatase‐1 (SULF1) in human bronchial epithelial cells (BECs) by crystalline silica (CS). In this study, CS‐induced down‐regulation of SULF1, and increases in Sulfated‐HSPGs, were determined in human BECs, and in rat lungs. By siRNA and plasmid transfection techniques the effects of SULF1 expression on silica‐induced fibrogenic and proliferative gene expression were determined. These studies confirmed down‐regulation of SULF1 and subsequent increases in sulfated‐HSPGs in vitro. Moreover, short‐term exposure of rats to CS resulted in similar changes in vivo. Conversely, effects were reversed after long term CS exposure of rats. SULF1 knockdown, and overexpression alleviated and exacerbated silica‐induced decrease in cell viability, respectively. Furthermore, overexpression of SULF1 promoted silica‐induced proliferative and fibrogenic gene expression, and collagen production. These findings demonstrate that the HSPG modification enzyme SULF1 and HSPG sulfation are altered by CS in vitro and in vivo. Furthermore, these changes may contribute to CS‐induced lung pathogenicity by affecting injury tolerance, hyperproliferation, and fibrotic effects. HighlightsThe 6‐O‐endosulfatase (Sulfatase‐1), is down‐regulated by exposure to crystalline‐silica in vitro and in vivo.Crystalline silica exposure alters heparin‐sulfate proteoglycan sulfation patterns in vitro and in vivo.Inhibition of Sulfatase‐1 expression has cytoprotective effects in human bronchial epithelial cells.Overexpression of Sulfatase‐1 exacerbates fibrogenic and hyperproliferative gene expression in bronchial epithelial cells.


European Respiratory Journal | 2014

Silica-induced inflammasome activation in vivo and uptake in vitro

Paul Peeters; Karin J. C. Sanders; Tim Perkins; Gernot Rohde; Emiel F.M. Wouters; Dirk Taeger; Beate Pesch; Niki L. Reynaert


European Respiratory Journal | 2013

Silica-induced inflammasome activation in vitro and in rat lungs

Paul Peeters; Niki L. Reynaert; Irene M. J. Eurlings; Timothy N Perkins; Brooke T. Mossman; Roel P. F. Schins; Emiel F. M. Wouters; Catrin Albrecht

Collaboration


Dive into the Paul Peeters's collaboration.

Top Co-Authors

Avatar

Niki L. Reynaert

Maastricht University Medical Centre

View shared research outputs
Top Co-Authors

Avatar

Emiel F.M. Wouters

Maastricht University Medical Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Catrin Albrecht

University of Düsseldorf

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ingrid Arijs

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Chad Steele

University of Alabama at Birmingham

View shared research outputs
Researchain Logo
Decentralizing Knowledge