Uilke Brouwer
University Medical Center Groningen
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Uilke Brouwer.
American Journal of Physiology-lung Cellular and Molecular Physiology | 2014
M. de Vries; Irene H. Heijink; R. Gras; L.E. den Boef; Marjan Reinders-Luinge; Simon D. Pouwels; Machteld N. Hylkema; M. van der Toorn; Uilke Brouwer; A. J. M. van Oosterhout; Martijn C. Nawijn
Exposure to cigarette smoke (CS) is the main risk factor for developing chronic obstructive pulmonary disease and can induce airway epithelial cell damage, innate immune responses, and airway inflammation. We hypothesized that cell survival factors might decrease the sensitivity of airway epithelial cells to CS-induced damage, thereby protecting the airways against inflammation upon CS exposure. Here, we tested whether Pim survival kinases could protect from CS-induced inflammation. We determined expression of Pim kinases in lung tissue, airway inflammation, and levels of keratinocyte-derived cytokine (KC) and several damage-associated molecular patterns in bronchoalveolar lavage in mice exposed to CS or air. Human bronchial epithelial BEAS-2B cells were treated with CS extract (CSE) in the presence or absence of Pim1 inhibitor and assessed for loss of mitochondrial membrane potential, induction of cell death, and release of heat shock protein 70 (HSP70). We observed increased expression of Pim1, but not of Pim2 and Pim3, in lung tissue after exposure to CS. Pim1-deficient mice displayed a strongly enhanced neutrophilic airway inflammation upon CS exposure compared with wild-type controls. Inhibition of Pim1 activity in BEAS-2B cells increased the loss of mitochondrial membrane potential and reduced cell viability upon CSE treatment, whereas release of HSP70 was enhanced. Interestingly, we observed release of S100A8 but not of double-strand DNA or HSP70 in Pim1-deficient mice compared with wild-type controls upon CS exposure. In conclusion, we show that expression of Pim1 protects against CS-induced cell death in vitro and neutrophilic airway inflammation in vivo. Our data suggest that the underlying mechanism involves CS-induced release of S100A8 and KC.
PLOS ONE | 2014
Akkelies E. Dijkstra; Joanna Smolonska; Maarten van den Berge; Ciska Wijmenga; Pieter Zanen; Marjan Luinge; Mathieu Platteel; Jan-Willem J. Lammers; Magnus Dahlbäck; Kerrie Tosh; Pieter S. Hiemstra; Peter J. Sterk; Avi Spira; Jørgen Vestbo; Børge G. Nordestgaard; Marianne Benn; Sune F. Nielsen; Morten Dahl; W. M. Monique Verschuren; H. Susan J. Picavet; Henriette A. Smit; Michael Owsijewitsch; Hans U. Kauczor; Harry J. de Koning; Eva Nizankowska-Mogilnicka; Filip Mejza; Pawel Nastalek; Cleo C. van Diemen; Michael H. Cho; Edwin K. Silverman
Background Chronic mucus hypersecretion (CMH) is associated with an increased frequency of respiratory infections, excess lung function decline, and increased hospitalisation and mortality rates in the general population. It is associated with smoking, but it is unknown why only a minority of smokers develops CMH. A plausible explanation for this phenomenon is a predisposing genetic constitution. Therefore, we performed a genome wide association (GWA) study of CMH in Caucasian populations. Methods GWA analysis was performed in the NELSON-study using the Illumina 610 array, followed by replication and meta-analysis in 11 additional cohorts. In total 2,704 subjects with, and 7,624 subjects without CMH were included, all current or former heavy smokers (≥20 pack-years). Additional studies were performed to test the functional relevance of the most significant single nucleotide polymorphism (SNP). Results A strong association with CMH, consistent across all cohorts, was observed with rs6577641 (p = 4.25×10−6, OR = 1.17), located in intron 9 of the special AT-rich sequence-binding protein 1 locus (SATB1) on chromosome 3. The risk allele (G) was associated with higher mRNA expression of SATB1 (4.3×10−9) in lung tissue. Presence of CMH was associated with increased SATB1 mRNA expression in bronchial biopsies from COPD patients. SATB1 expression was induced during differentiation of primary human bronchial epithelial cells in culture. Conclusions Our findings, that SNP rs6577641 is associated with CMH in multiple cohorts and is a cis-eQTL for SATB1, together with our additional observation that SATB1 expression increases during epithelial differentiation provide suggestive evidence that SATB1 is a gene that affects CMH.
American Journal of Physiology-lung Cellular and Molecular Physiology | 2015
Simon D. Pouwels; Irene H. Heijink; Uilke Brouwer; R. Gras; Lisette E. den Boef; H. Marike Boezen; Ron Korstanje; Antoon J. M. van Oosterhout; Martijn C. Nawijn
Neutrophilic airway inflammation is one of the major hallmarks of chronic obstructive pulmonary disease and is also seen in steroid resistant asthma. Neutrophilic airway inflammation can be induced by different stimuli including cigarette smoke (CS). Short-term exposure to CS induces neutrophilic airway inflammation in both mice and humans. Since not all individuals develop extensive neutrophilic airway inflammation upon smoking, we hypothesized that this CS-induced innate inflammation has a genetic component. This hypothesis was addressed by exposing 30 different inbred mouse strains to CS or control air for 5 consecutive days, followed by analysis of neutrophilic lung inflammation. By genomewide haplotype association mapping, we identified four susceptibility genes with a significant association to lung tissue levels of the neutrophil marker myeloperoxidase under basal conditions and an additional five genes specifically associated with CS-induced tissue MPO levels. Analysis of the expression levels of the susceptibility genes by quantitative RT-PCR revealed that three of the four genes associated with CS-induced tissue MPO levels had CS-induced changes in gene expression levels that correlate with CS-induced airway inflammation. Most notably, CS exposure induces an increased expression of the coiled-coil domain containing gene, Ccdc93, in mouse strains susceptible for CS-induced airway inflammation whereas Ccdc93 expression was decreased upon CS exposure in nonsusceptible mouse strains. In conclusion, this study shows that CS-induced neutrophilic airway inflammation has a genetic component and that several genes contribute to the susceptibility for this response.
American Journal of Physiology-lung Cellular and Molecular Physiology | 2015
Grissel Faura Tellez; Karl Vandepoele; Uilke Brouwer; Henk Koning; Robin M. Elderman; Tillie-Louise Hackett; Brigitte Willemse; John W. Holloway; Frans van Roy; Gerard H. Koppelman; Martijn C. Nawijn
Genetic studies have identified Protocadherin-1 (PCDH1) and Mothers against decapentaplegic homolog-3 (SMAD3) as susceptibility genes for asthma. PCDH1 is expressed in bronchial epithelial cells and has been found to interact with SMAD3 in yeast two-hybrid (Y2H) overexpression assays. Here, we test whether PCDH1 and SMAD3 interact at endogenous protein levels in bronchial epithelial cells and evaluate the consequences thereof for transforming growth factor-β1 (TGF-β1)-induced gene transcription. We performed Y2H screens and coimmunoprecipitation (co-IP) experiments of PCDH1 and SMAD3 in HEK293T and 16HBE14o(-) (16HBE) cell lines. Activity of a SMAD3-driven luciferase reporter gene in response to TGF-β1 was measured in BEAS-2B cells transfected with PCDH1 and in 16HBE cells transfected with PCDH1-small-interfering RNA (siRNA). TGF-β1-induced gene expression was quantified in BEAS-2B clones overexpressing PCDH1 and in human primary bronchial epithelial cells (PBECs) transfected with PCDH1-siRNA. We confirm PCDH1 and SMAD3 interactions by Y2H and by co-IP in HEK293T cells overexpressing both proteins, and at endogenous protein levels in 16HBE cells. TGF-β-induced activation of a SMAD3-driven reporter was reduced by exogenous PCDH1 in BEAS2B cells, whereas it was increased by siRNA-mediated knockdown of endogenous PCDH1 in 16HBE cells. Overexpression of PCDH1 suppressed expression of TGF-β target genes in BEAS-2B cells, whereas knockdown of PCDH1 in human PBECs increased TGF-β-induced gene expression. In conclusion, we demonstrate that PCDH1 binds to SMAD3 and regulates its activation by TGF-β signaling in bronchial epithelial cells. We propose that PCDH1 and SMAD3 act in a single pathway in asthma susceptibility that affects sensitivity of the airway epithelium to TGF-β.
PLOS ONE | 2014
Henk Koning; Antoon J. M. van Oosterhout; Uilke Brouwer; Lisette E. den Boef; R. Gras; Marjan Reinders-Luinge; Corry-Anke Brandsma; Marco van der Toorn; Machteld N. Hylkema; Brigitte Willemse; Ian Sayers; Gerard H. Koppelman; Martijn C. Nawijn
Protocadherin-1 (PCDH1) is a novel susceptibility gene for airway hyperresponsiveness, first identified in families exposed to cigarette smoke and is expressed in bronchial epithelial cells. Here, we asked how mouse Pcdh1 expression is regulated in lung structural cells in vivo under physiological conditions, and in both short-term cigarette smoke exposure models characterized by airway inflammation and hyperresponsiveness and chronic cigarette smoke exposure models. Pcdh1 gene-structure was investigated by Rapid Amplification of cDNA Ends. Pcdh1 mRNA and protein expression was investigated by qRT-PCR, western blotting using isoform-specific antibodies. We observed 87% conservation of the Pcdh1 nucleotide sequence, and 96% conservation of the Pcdh1 protein sequence between men and mice. We identified a novel Pcdh1 isoform encoding only the intracellular signalling motifs. Cigarette smoke exposure for 4 consecutive days markedly reduced Pcdh1 mRNA expression in lung tissue (3 to 4-fold), while neutrophilia and airway hyperresponsiveness was induced. Moreover, Pcdh1 mRNA expression in lung tissue was reduced already 6 hours after an acute cigarette-smoke exposure in mice. Chronic exposure to cigarette smoke induced loss of Pcdh1 protein in lung tissue after 2 months, while Pcdh1 protein levels were no longer reduced after 9 months of cigarette smoke exposure. We conclude that Pcdh1 is highly homologous to human PCDH1, encodes two transmembrane proteins and one intracellular protein, and is regulated by cigarette smoke exposure in vivo.
PLOS ONE | 2016
Grissel Faura Tellez; Brigitte Willemse; Uilke Brouwer; Susan Nijboer-Brinksma; Karl Vandepoele; Jacobien A. Noordhoek; Irene H. Heijink; Maaike de Vries; Natalie P. Smithers; Dirkje S. Postma; Wim Timens; Laura Wiffen; Frans van Roy; John W. Holloway; Peter M. Lackie; Martijn C. Nawijn; Gerard H. Koppelman
Background The asthma gene PCDH1 encodes Protocadherin-1, a putative adhesion molecule of unknown function expressed in the airway epithelium. Here, we characterize the localization, differential expression, homotypic adhesion specificity and function of PCDH1 in airway epithelial cells in asthma. Methods We performed confocal fluorescence microscopy to determine subcellular localization of PCDH1 in 16HBE cells and primary bronchial epithelial cells (PBECs) grown at air-liquid interface. Next, to compare PCDH1 expression and localization in asthma and controls we performed qRT-PCR and fluorescence microscopy in PBECs and immunohistochemistry on airway wall biopsies. We examined homotypic adhesion specificity of HEK293T clones overexpressing fluorescently tagged-PCDH1 isoforms. Finally, to evaluate the role for PCDH1 in epithelial barrier formation and repair, we performed siRNA knockdown-studies and measured epithelial resistance. Results PCDH1 localized to the cell membrane at cell-cell contact sites, baso-lateral to adherens junctions, with increasing expression during epithelial differentiation. No differences in gene expression or localization of PCDH1 isoforms expressing the extracellular domain were observed in either PBECs or airway wall biopsies between asthma patients and controls. Overexpression of PCDH1 mediated homotypic interaction, whereas downregulation of PCDH1 reduced epithelial barrier formation, and impaired repair after wounding. Conclusions In conclusion, PCDH1 is localized to the cell membrane of bronchial epithelial cells baso-lateral to the adherens junction. Expression of PCDH1 is not reduced nor delocalized in asthma even though PCDH1 contributes to homotypic adhesion, epithelial barrier formation and repair.
Clinical & Experimental Allergy | 2018
Laura Hesse; Uilke Brouwer; Arjen H. Petersen; R. Gras; L. Bosman; J. Brimnes; J. N. G. Oude Elberink; A. J. M. van Oosterhout; Martijn C. Nawijn
Both subcutaneous and sublingual allergen immunotherapy (SCIT and SLIT) have been shown to effectively suppress allergic manifestations upon allergen exposure, providing long‐term relief from symptoms in allergic disorders including allergic asthma. Clinical studies directly comparing SCIT and SLIT report a different kinetics and magnitude of immunological changes induced during treatment. Comparative studies into the mechanisms underlying immune suppression in SCIT and SLIT are lacking.
PLOS ONE | 2015
Akkelies E. Dijkstra; Joanna Smolonska; Maarten van den Berge; Ciska Wijmenga; Pieter Zanen; Marjan Luinge; Mathieu Platteel; Jan-Willem J. Lammers; Magnus Dahlbäck; Kerrie Tosh; Pieter S. Hiemstra; Peter J. Sterk; Avi Spira; Jørgen Vestbo; Børge G. Nordestgaard; Marianne Benn; Sune F. Nielsen; Morten Dahl; W. M. Monique Verschuren; H. Susan J. Picavet; Henriette A. Smit; Michael Owsijewitsch; Hans U. Kauczor; Harry J. de Koning; Eva Nizankowska-Mogilnicka; Filip Mejza; Pawel Nastalek; Cleo C. van Diemen; Michael H. Cho; Edwin K. Silverman
Akkelies E. Dijkstra, Joanna Smolonska, Maarten van den Berge, Ciska Wijmenga, Pieter Zanen, Marjan A. Luinge, Mathieu Platteel, Jan-Willem Lammers, Magnus Dahlback, Kerrie Tosh, Pieter S. Hiemstra, Peter J. Sterk, Avi Spira, Jorgen Vestbo, Borge G. Nordestgaard, Marianne Benn, Sune F. Nielsen, Morten Dahl, W. Monique Verschuren, H. Susan J. Picavet, Henriette A. Smit, Michael Owsijewitsch, Hans U. Kauczor, Harry J. de Koning, Eva Nizankowska-Mogilnicka, Filip Mejza, Pawel Nastalek, Cleo C. van Diemen, Michael H. Cho, Edwin K. Silverman, James D. Crapo, Terri H. Beaty, David A. Lomas, Per Bakke, Amund Gulsvik, Yohan Bosse, Ma’en Obeidat, DaanW. Loth, Lies Lahousse, Fernando Rivadeneira, Andre G. Uitterlinden, Andre Hofman, Bruno H. Stricker, Guy G. Brusselle, Cornelia M. van Duijn, Uilke Brouwer, Gerard H. Koppelman, Judith M. Vonk, Martijn C. Nawijn, Harry J. M. Groen, Wim Timens, H. Marike Boezen, Dirkje S. Postma, the LifeLines Cohort study
PLOS ONE | 2015
Akkelies E. Dijkstra; Joanna Smolonska; Maarten van den Berge; Ciska Wijmenga; Pieter Zanen; Marjan Luinge; Mathieu Platteel; Jan-Willem J. Lammers; Magnus Dahlbäck; Kerrie Tosh; Pieter S. Hiemstra; Peter J. Sterk; Avi Spira; Jørgen Vestbo; Børge G. Nordestgaard; Marianne Benn; Sune F. Nielsen; Morten Dahl; W. M. Monique Verschuren; H. Susan J. Picavet; Henriette A. Smit; Michael Owsijewitsch; Hans U. Kauczor; Harry J. de Koning; Eva Nizankowska-Mogilnicka; Filip Mejza; Pawel Nastalek; Cleo C. van Diemen; Michael H. Cho; Edwin K. Silverman
European Respiratory Journal | 2015
Grissel Faura Tellez; Uilke Brouwer; Brigitte Willemse; Jacobien A. Noordhoek; Irene Hejink; Maaike Vries de; Natalie P. Smithers; Dirkje S. Postma; Wim Timens; Laura Wiffen; John W. Holloway; Peter M. Lackie; Martijn C. Nawijn; Gerard H. Koppelman