Network


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

Hotspot


Dive into the research topics where Fien M. Verhamme is active.

Publication


Featured researches published by Fien M. Verhamme.


American Journal of Respiratory and Critical Care Medicine | 2013

Role of CXCL13 in Cigarette Smoke–induced Lymphoid Follicle Formation and Chronic Obstructive Pulmonary Disease

Ken R. Bracke; Fien M. Verhamme; Leen Seys; Claudie Bantsimba-Malanda; Danen Cunoosamy; Ronald Herbst; Hamida Hammad; Bart N. Lambrecht; Guy Joos; Guy Brusselle

RATIONALE The B cell-attracting chemokine CXCL13 is an important mediator in the formation of tertiary lymphoid organs (TLOs). Increased numbers of ectopic lymphoid follicles have been observed in lungs of patients with severe chronic obstructive pulmonary disease (COPD). However, the role of these TLOs in the pathogenesis of COPD remains unknown. OBJECTIVES By neutralizing CXCL13 in a mouse model of chronic cigarette smoke (CS) exposure, we aimed at interrogating the link between lymphoid follicles and development of pulmonary inflammation, emphysema, and airway wall remodeling. METHODS We first quantified and localized CXCL13 in lungs of air- or CS-exposed mice and in lungs of never smokers, smokers without airflow obstruction, and patients with COPD by reverse transcriptase-polymerase chain reaction, ELISA, and immunohistochemistry. Next, CXCL13 signaling was blocked by prophylactic or therapeutic administration of anti-CXCL13 antibodies in mice exposed to air or CS for 24 weeks, and several hallmarks of COPD were evaluated. MEASUREMENTS AND MAIN RESULTS Both mRNA and protein levels of CXCL13 were increased in lungs of CS-exposed mice and patients with COPD. Importantly, expression of CXCL13 was observed within B-cell areas of lymphoid follicles. Prophylactic and therapeutic administration of anti-CXCL13 antibodies completely prevented the CS-induced formation of pulmonary lymphoid follicles in mice. Interestingly, absence of TLOs attenuated destruction of alveolar walls and inflammation in bronchoalveolar lavage but did not affect airway wall remodeling. CONCLUSIONS CXCL13 is produced within lymphoid follicles of patients with COPD and is crucial for the formation of TLOs. Neutralization of CXCL13 partially protects mice against CS-induced inflammation in bronchoalveolar lavage and alveolar wall destruction.


PLOS ONE | 2016

Characterization and Quantification of Innate Lymphoid Cell Subsets in Human Lung

Katrien De Grove; Sharen Provoost; Fien M. Verhamme; Ken R. Bracke; Guy Joos; Tania Maes; Guy Brusselle

Background Innate lymphoid cells (ILC) are a new family of innate immune cells that have emerged as important regulators of tissue homeostasis and inflammation. However, limited data are available concerning the relative abundance and characteristics of ILC in the human lung. Methods The aim of this study was to characterize and enumerate the different ILC subsets in human lung by multi-color flow cytometry. Results Within the CD45+ Lin- CD127+ pulmonary ILC population, we identified group 1 (ILC1), group 2 (ILC2) and group 3 (ILC3) innate lymphoid cells using specific surface markers (i.e. IL12Rβ2, CRTH2 and CD117 respectively) and key transcription factors (i.e. T-bet, GATA-3 and RORγT respectively). Based on the presence of NKp44, ILC3 were further subdivided in natural cytotoxicity receptor (NCR)+ and NCR- ILC3. In addition, we demonstrated the production of signature cytokines IFN-γ, IL-5, IL-17A, IL-22 and GM-CSF in the pulmonary ILC population. Interestingly, we observed a tendency to a higher frequency of NCR- ILC3 in lungs of patients with chronic obstructive pulmonary disease (COPD) compared with controls. Conclusions We show that the three main ILC subsets are present in human lung. Importantly, the relative abundance of ILC subsets tended to change in COPD patients in comparison to control individuals.


American Journal of Respiratory and Critical Care Medicine | 2015

Role of B Cell-Activating Factor in Chronic Obstructive Pulmonary Disease.

Leen Seys; Fien M. Verhamme; Anja Schinwald; Hamida Hammad; Danen Cunoosamy; Claudie Bantsimba-Malanda; Alan Sabirsh; Eileen McCall; Liz Flavell; Ronald Herbst; Sharen Provoost; Bart N. Lambrecht; Guy Joos; Guy Brusselle; Ken R. Bracke

RATIONALE B cell-activating factor (BAFF) plays a major role in activation of B cells and in adaptive humoral immune responses. In chronic obstructive pulmonary disease (COPD), lymphoid follicles have been associated with disease severity, and overexpression of BAFF has been demonstrated within lymphoid follicles of patients with severe COPD. OBJECTIVES To investigate expression and localization of BAFF in the lungs of patients with COPD and to study the role of BAFF in COPD by antagonizing BAFF in a mouse model of chronic cigarette smoke (CS) exposure. METHODS We quantified and localized BAFF expression in lungs of never-smokers, smokers without COPD, and patients with COPD and in lungs of air- or CS-exposed mice by reverse-transcriptase polymerase chain reaction, ELISA, immunohistochemistry, and confocal imaging. Next, to investigate the role of BAFF in COPD, we antagonized BAFF by prophylactic or therapeutic administration of a soluble fusion protein of the BAFF-receptor, BAFFR-Fc, in mice exposed to air or CS for 24 weeks and evaluated several hallmarks of COPD and polarization of lung macrophages. MEASUREMENTS AND MAIN RESULTS BAFF expression was significantly increased in lungs of patients with COPD and CS-exposed mice. BAFF staining in lymphoid follicles was observed around B cells, CD4(+) cells, dendritic cells, follicular dendritic cells, and fibroblastic reticular cells. Prophylactic and therapeutic administration of BAFFR-Fc in mice reduced pulmonary B-cell numbers and prevented CS-induced formation of lymphoid follicles and increases in immunoglobulin levels. Interestingly, prophylactic BAFFR-Fc administration significantly attenuated pulmonary inflammation and destruction of alveolar walls. Moreover, antagonizing BAFF altered the phenotype of alveolar and interstitial macrophages. CONCLUSIONS BAFF is significantly increased in lungs of patients with COPD and is present around both immune and stromal cells within lymphoid follicles. Antagonizing BAFF in CS-exposed mice attenuates pulmonary inflammation and alveolar destruction.


American Journal of Respiratory and Critical Care Medicine | 2017

MicroRNA Profiling Reveals a Role for MicroRNA-218-5p in the Pathogenesis of Chronic Obstructive Pulmonary Disease

Griet Conickx; Pieter Mestdagh; Avila Cobos F; Fien M. Verhamme; Tania Maes; Bart Vanaudenaerde; Leen Seys; Lies Lahousse; R.Y. Kim; Alan Hsu; Peter Wark; Philip M. Hansbro; Guy Joos; Jo Vandesompele; Ken R. Bracke; Guy Brusselle

Rationale: Aberrant expression of microRNAs (miRNAs) can have a detrimental role in disease pathogenesis. Objectives: To identify dysregulated miRNAs in lung tissue of patients with chronic obstructive pulmonary disease (COPD). Methods: We performed miRNA and mRNA profiling using high throughput stem‐loop reverse‐transcriptase quantitative polymerase chain reaction and mRNA microarray, respectively, on lung tissue of 30 patients (screening cohort) encompassing 8 never‐smokers, 10 smokers without airflow limitation, and 12 smokers with COPD. Differential expression of miRNA‐218‐5p (miR‐218‐5p) was validated by reverse‐transcriptase quantitative polymerase chain reaction in an independent cohort of 71 patients, an in vivo murine model of COPD, and primary human bronchial epithelial cells. Localization of miR‐218‐5p was assessed by in situ hybridization. In vitro and in vivo perturbation of miR‐218‐5p combined with RNA sequencing and gene set enrichment analysis was used to elucidate its functional role in COPD pathogenesis. Measurements and Main Results: Several miRNAs were differentially expressed among the different patient groups. Interestingly, miR‐218‐5p was significantly down‐regulated in smokers without airflow limitation and in patients with COPD compared with never‐smokers. Decreased pulmonary expression of miR‐218‐5p was validated in an independent validation cohort, in cigarette smoke‐exposed mice, and in human bronchial epithelial cells. Importantly, expression of miR‐218‐5p strongly correlated with airway obstruction. Furthermore, cellular localization of miR‐218‐5p in human and murine lung revealed highest expression of miR‐218‐5p in the bronchial airway epithelium. Perturbation experiments with a miR‐218‐5p mimic or inhibitor demonstrated a protective role of miR‐218‐5p in cigarette smoke‐induced inflammation and COPD. Conclusions: We highlight a role for miR‐218‐5p in the pathogenesis of COPD.


European Respiratory Journal | 2014

Role of activin-A in cigarette smoke-induced inflammation and COPD.

Fien M. Verhamme; Ken R. Bracke; Gimano D. Amatngalim; Geert Verleden; Geert R. Van Pottelberge; Pieter S. Hiemstra; Guy Joos; Guy Brusselle

Activin-A is a pleiotropic cytokine belonging to the transforming growth factor-&bgr; superfamily and has been implicated in asthma and pulmonary fibrosis. However, the role of activin-A and its endogenous inhibitor, follistatin, in the pathogenesis of chronic obstructive pulmonary disease (COPD) is unknown. We first quantified activin-A and follistatin in the lungs of air- or cigarette smoke-exposed mice and in the lungs of patients with COPD by immunohistochemistry, ELISA and quantitative real-time PCR. We subsequently studied the effect of cigarette smoke on primary human bronchial epithelial cells in vitro. Next, activin-A signalling was antagonised in vivo by administration of follistatin in mice exposed to air or cigarette smoke for 4 weeks. Protein levels of activin-A were increased in the airway epithelium of patients with COPD compared with never-smokers and smokers. Cigarette smoke-exposed human bronchial epithelial cells expressed higher levels of activin-A and lower levels of follistatin. Both mRNA and protein levels of activin-A were increased in the lungs of cigarette smoke-exposed mice, whereas follistatin levels were reduced upon cigarette smoke exposure. Importantly, administration of follistatin attenuated the cigarette smoke-induced increase of inflammatory cells and mediators in the bronchoalveolar lavage fluid in mice. These results suggest that an imbalance between activin-A and follistatin contributes to the pathogenesis of cigarette smoke-induced inflammation and COPD. Imbalance of activin-A and FST in favour of activin-A signalling in COPD patients cigarette smoke-exposed mice http://ow.ly/qMqVN


PLOS ONE | 2014

Cigarette Smoke Extract Induces a Phenotypic Shift in Epithelial Cells; Involvement of HIF1α in Mesenchymal Transition

Irene M. J. Eurlings; Niki L. Reynaert; Twan van den Beucken; Harry R. Gosker; C. C. de Theije; Fien M. Verhamme; Ken R. Bracke; Emiel F. M. Wouters; Mieke A. Dentener

In COPD, matrix remodeling contributes to airflow limitation. Recent evidence suggests that next to fibroblasts, the process of epithelial-mesenchymal transition can contribute to matrix remodeling. CSE has been shown to induce EMT in lung epithelial cells, but the signaling mechanisms involved are largely unknown and subject of this study. EMT was assessed in A549 and BEAS2B cells stimulated with CSE by qPCR, Western blotting and immunofluorescence for epithelial and mesenchymal markers, as were collagen production, cell adhesion and barrier integrity as functional endpoints. Involvement of TGF-β and HIF1α signaling pathways were investigated. In addition, mouse models were used to examine the effects of CS on hypoxia signaling and of hypoxia per se on mesenchymal expression. CSE induced EMT characteristics in A549 and BEAS2B cells, evidenced by decreased expression of epithelial markers and a concomitant increase in mesenchymal marker expression after CSE exposure. Furthermore cells that underwent EMT showed increased production of collagen, decreased adhesion and disrupted barrier integrity. The induction of EMT was found to be independent of TGF-β signaling. On the contrary, CS was able to induce hypoxic signaling in A549 and BEAS2B cells as well as in mice lung tissue. Importantly, HIF1α knock-down prevented induction of mesenchymal markers, increased collagen production and decreased adhesion after CSE exposure, data that are in line with the observed induction of mesenchymal marker expression by hypoxia in vitro and in vivo. Together these data provide evidence that both bronchial and alveolar epithelial cells undergo a functional phenotypic shift in response to CSE exposure which can contribute to increased collagen deposition in COPD lungs. Moreover, HIF1α signaling appears to play an important role in this process.


American Journal of Respiratory Cell and Molecular Biology | 2015

Transforming Growth Factor-β Superfamily in Obstructive Lung Diseases. More Suspects Than TGF-β Alone

Fien M. Verhamme; Ken R. Bracke; Guy Joos; Guy Brusselle

Asthma and chronic obstructive pulmonary disease are respiratory disorders and a major global health problem with increasing incidence and severity. Genes originally associated with lung development could be relevant in the pathogenesis of chronic obstructive pulmonary disease/asthma, owing to either an early-life origin of adult complex diseases or their dysregulation in adulthood upon exposure to environmental stressors (e.g., smoking). The transforming growth factor (TGF)-β superfamily is conserved through evolution and is involved in a range of biological processes, both during development and in adult tissue homeostasis. TGF-β1 has emerged as an important regulator of lung and immune system development. However, considerable evidence has been presented for a role of many of the other ligands of the TGF-β superfamily in lung pathology, including activins, bone morphogenetic proteins, and growth differentiation factors. In this review, we summarize the current knowledge on the mechanisms by which activin, bone morphogenetic protein, and growth differentiation factor signaling contribute to the pathogenesis of obstructive airway diseases.


PLOS ONE | 2015

Airway Surface Dehydration Aggravates Cigarette Smoke-Induced Hallmarks of COPD in Mice.

Leen Seys; Fien M. Verhamme; Lisa L. Dupont; Elke Desauter; Julia Duerr; Ayca Seyhan Agircan; Griet Conickx; Guy Joos; Guy Brusselle; Marcus A. Mall; Ken R. Bracke

Introduction Airway surface dehydration, caused by an imbalance between secretion and absorption of ions and fluid across the epithelium and/or increased epithelial mucin secretion, impairs mucociliary clearance. Recent evidence suggests that this mechanism may be implicated in chronic obstructive pulmonary disease (COPD). However, the role of airway surface dehydration in the pathogenesis of cigarette smoke (CS)-induced COPD remains unknown. Objective We aimed to investigate in vivo the effect of airway surface dehydration on several CS-induced hallmarks of COPD in mice with airway-specific overexpression of the β-subunit of the epithelial Na+ channel (βENaC). Methods βENaC-Tg mice and wild-type (WT) littermates were exposed to air or CS for 4 or 8 weeks. Pathological hallmarks of COPD, including goblet cell metaplasia, mucin expression, pulmonary inflammation, lymphoid follicles, emphysema and airway wall remodelling were determined and lung function was measured. Results Airway surface dehydration in βENaC-Tg mice aggravated CS-induced airway inflammation, mucin expression and destruction of alveolar walls and accelerated the formation of pulmonary lymphoid follicles. Moreover, lung function measurements demonstrated an increased compliance and total lung capacity and a lower resistance and hysteresis in βENaC-Tg mice, compared to WT mice. CS exposure further altered lung function measurements. Conclusions We conclude that airway surface dehydration is a risk factor that aggravates CS-induced hallmarks of COPD.


Molecular and Cellular Endocrinology | 2017

Increased levels of enzymes involved in local estradiol synthesis in chronic obstructive pulmonary disease

Gonda Konings; Niki L. Reynaert; Bert Delvoux; Fien M. Verhamme; Ken R. Bracke; Guy Brusselle; Andrea Romano; Juanita H. J. Vernooy

INTRODUCTION Steroid hormones are involved in lung development, pulmonary inflammation, and lung cancer. Estrogen signaling and exposure may play a role in pulmonary disorders, including COPD. In both genders, estrogens can be generated locally in the lungs and this contributes importantly to the tissue exposure to these steroids. OBJECTIVE To characterize and assess differences in localization of estrogen receptors and enzymes involved in the local generation of estrogens in COPD. METHODS Estrogen Receptor alpha (ERα/ESR1), Estrogen Receptor beta (ERβ/ESR2) and G-protein-coupled estrogen receptor 1 (GPER) were explored by real-time (RT)-PCR analysis (mRNA expression), immunohistochemistry and western blotting in controls and COPD patients. mRNA expression of the enzymes involved in the local estrogen generation - i.e. aromatase (CYP19A1), 17beta-hydroxysteroid dehydrogenases (17β-HSDs) 1, 2, 4, 5, 7 and 12, steroid sulfatase (STS) and sulfotransferase (SULT1E1) - were analyzed by RT-PCR. RESULTS ERα, ERβ and GPER were expressed in lung tissue, but no differences were observed between patients and controls. The main enzymes involved in local estrogen generation were also present in both normal and COPD lung tissue. In lungs of COPD patients compared with controls, we observed increased expression of the enzymes 17β-HSD type 1 and aromatase (positive association), both involved in the local synthesis of active estrogens. CONCLUSION All ER subtypes are present in the lung. The shift in local mRNA level of estrogen metabolic enzymes suggests that exposure to estrogens is involved in the pathogenesis of COPD.


Clinical Infectious Diseases | 2018

DPP4, the Middle East Respiratory Syndrome Coronavirus Receptor, is Upregulated in Lungs of Smokers and Chronic Obstructive Pulmonary Disease Patients

Leen Seys; W. Widagdo; Fien M. Verhamme; Alex KleinJan; Wim Janssens; Guy Joos; Ken R. Bracke; Bart L. Haagmans; Guy Brusselle

The MERS coronavirus receptor, dipeptidyl peptidase 4 is upregulated in the lungs of smokers and chronic obstructive pulmonary disease (COPD) patients. This offers a potential mechanism for the increased susceptibility of smokers and COPD patients for MERS-CoV infection.

Collaboration


Dive into the Fien M. Verhamme's collaboration.

Top Co-Authors

Avatar

Ken R. Bracke

Ghent University Hospital

View shared research outputs
Top Co-Authors

Avatar

Guy Brusselle

Ghent University Hospital

View shared research outputs
Top Co-Authors

Avatar

Guy Joos

Ghent University Hospital

View shared research outputs
Top Co-Authors

Avatar

Leen Seys

Ghent University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bart Vanaudenaerde

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Hamida Hammad

Flanders Institute for Biotechnology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sharen Provoost

Ghent University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge