Network


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

Hotspot


Dive into the research topics where Katrin Hostettler is active.

Publication


Featured researches published by Katrin Hostettler.


European Respiratory Journal | 2015

Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis

Lutz Wollin; Eva Wex; Alexander Pautsch; Gisela Schnapp; Katrin Hostettler; Susanne Stowasser; Martin Kolb

Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal disease characterised by fibrosis of the lung parenchyma and loss of lung function. Although the pathogenic pathways involved in IPF have not been fully elucidated, IPF is believed to be caused by repetitive alveolar epithelial cell injury and dysregulated repair, in which there is uncontrolled proliferation of lung fibroblasts and differentiation of fibroblasts into myofibroblasts, which excessively deposit extracellular matrix (ECM) proteins in the interstitial space. A number of profibrotic mediators including platelet-derived growth factor (PDGF), fibroblast growth factor (FGF) and transforming growth factor-β are believed to play important roles in the pathogenesis of IPF. Nintedanib is a potent small molecule inhibitor of the receptor tyrosine kinases PDGF receptor, FGF receptor and vascular endothelial growth factor receptor. Data from in vitro studies have shown that nintedanib interferes with processes active in fibrosis such as fibroblast proliferation, migration and differentiation, and the secretion of ECM. In addition, nintedanib has shown consistent anti-fibrotic and anti-inflammatory activity in animal models of lung fibrosis. These data provide a strong rationale for the clinical efficacy of nintedanib in patients with IPF, which has recently been demonstrated in phase III clinical trials. Nintedanib interferes with processes active in fibrosis, e.g. fibroblast proliferation, migration and differentiation http://ow.ly/Iae9z


Transplantation | 2004

Everolimus and mycophenolate mofetil are potent inhibitors of fibroblast proliferation after lung transplantation.

Andrea Azzola; A. Havryk; Prashant N. Chhajed; Katrin Hostettler; Judith L. Black; Peter R. A. Johnson; Michael Roth; Allan R. Glanville; Michael Tamm

Background. Dysregulated fibroblast proliferation is thought to play an important role in the progression of bronchiolitis obliterans (BO) after lung transplantation. Augmented immunosuppression is often used to treat BO. We investigated the effect of methylprednisolone (mPRED), cyclosporine A (CsA), tacrolimus (FK506), azathioprine (AZA), mycophenolate mofetil (MMF), and everolimus (rapamycin derivative [RAD]) on the proliferative capacity of fibroblasts cultured from transbronchial biopsies of lung transplant recipients. Methods. Primary cultures of human lung fibroblasts were obtained from 14 transbronchial biopsies of lung transplant recipients. Subconfluent cells were serum starved for 24 hr followed by growth stimulation in the presence or absence of the respective drug in six concentrations ranging as follows: 0.01 to 100 mg/L for mPRED; 0.01 to 50 mg/L for CsA and AZA; 0.001 to 5 mg/L for FK506 and MMF; and 0.00001 to 1 mg/L for RAD. Proliferation was quantified by [3H]thymidine incorporation and direct cell count. A toxic drug effect was excluded by trypan blue. Results. Drug concentrations (mg/L) causing a 50% inhibition of fibroblast proliferation were mPRED 4; CsA 20; FK506 0.3; AZA 7; MMF 0.3; and RAD 0.0006. Drug concentrations (mg/L) causing inhibition of fetal bovine serum-induced proliferation were mPRED 60; CsA 45; FK506 3; AZA 35; MMF 1; and RAD 0.003. Conclusions. RAD and MMF were the most potent antifibroproliferative drugs and were effective at concentrations achieved clinically, supporting their use for the treatment of patients with early BO. Our method holds promise as an in vitro model to assess the likely in vivo responses of human lung fibroblasts to specific immunosuppressive drugs.


Respiratory Research | 2014

Anti-fibrotic effects of nintedanib in lung fibroblasts derived from patients with idiopathic pulmonary fibrosis

Katrin Hostettler; Jun Zhong; Eleni Papakonstantinou; George Karakiulakis; Michael Tamm; Petra Seidel; Qingzhu Sun; Jyotshna Mandal; Didier Lardinois; Christopher Lambers; Michael Roth

BackgroundIdiopathic pulmonary fibrosis (IPF) is a progressive lung disease with poor prognosis. The kinase inhibitor nintedanib specific for vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) and fibroblast growth factor receptor (FGFR) significantly reduced the rate of decline of forced vital capacity versus placebo.AimTo determine the in vitro effect of nintedanib on primary human lung fibroblasts. Methods: Fibroblasts were isolated from lungs of IPF patients and from non-fibrotic controls. We assessed the effect of VEGF, PDGF-BB and basic FGF (bFGF) ± nintedanib on: (i) expression/activation of VEGFR, PDGFR, and FGFR, (ii) cell proliferation, secretion of (iii) matrix metalloproteinases (MMP), (iv) tissue inhibitor of metalloproteinase (TIMP), and (v) collagen.ResultsIPF fibroblasts expressed higher levels of PDGFR and FGFR than controls. PDGF-BB, bFGF, and VEGF caused a pro-proliferative effect which was prevented by nintedanib. Nintedanib enhanced the expression of pro-MMP-2, and inhibited the expression of TIMP-2. Transforming growth factor-beta-induced secretion of collagens was inhibited by nintedanib.ConclusionOur data demonstrate a significant anti-fibrotic effect of nintedanib in IPF fibroblasts. This effect consists of the drug’s anti-proliferative capacity, and on its effect on the extracellular matrix, the degradation of which seems to be enhanced.


Respiration | 2012

Long-term treatment with infliximab in patients with sarcoidosis.

Katrin Hostettler; Ueli Studler; Michael Tamm; Martin Brutsche

Background: Long-term benefit and safety of infliximab treatment in patients with chronic sarcoidosis remain unclear. Objectives: It was the aim of this study to assess the clinical benefit and safety of long-term infliximab treatment in patients with chronic steroid-resistant sarcoidosis. Methods: We conducted a retrospective chart review of all patients with chronic steroid-resistant sarcoidosis who received infliximab between January 2003 and November 2010. Pulmonary function tests and index lesions before and after infliximab therapy were assessed. Results: Between January 2003 and November 2010, 28 patients received in-fliximab, 16 of them for more than 12 months. Five (31%) of these 16 patients with long-term infliximab treatment had a predominantly pulmonary disease, whereas 11 (69%) had a predominantly extrapulmonary involvement. Mean duration of treatment for the 16 patients was 29 months (range 12-62). Six of 11 (55%) patients with mainly extrapulmonary sarcoidosis showed a complete remission of their index lesion, 4/11 (36%) had a partial remission and 1/11 (9%) showed no response. One out of 5 patients with predominantly pulmonary sarcoidosis showed a >10% improvement in percentage predicted forced vital capacity, 3/5 showed a 0-10% improvement, and in 1/5 patients, percentage predicted forced vital capacity declined during infliximab treatment. Thus, overall, 14/16 (88%) patients profited from long-term infliximab treatment. Suspected adverse effects which lead to a temporary withdrawal of infliximab therapy were noticed in 1/16 (6%) patients. Conclusions: This retrospective study indicates that long-term infliximab is very efficient and safe in patients with chronic steroid-resistant sarcoidosis when assessed with individualized treatment targets. Patients with predominantly extrapulmonary sarcoidosis seem to profit more than patients with a predominantly pulmonary disease.


PLOS ONE | 2013

HGF Expressing Stem Cells in Usual Interstitial Pneumonia Originate from the Bone Marrow and Are Antifibrotic

Amiq Gazdhar; Njomeza Susuri; Katrin Hostettler; Mathias Gugger; Lars Knudsen; Michael Roth; Matthias Ochs; Thomas Geiser

Background Pulmonary fibrosis may result from abnormal alveolar wound repair after injury. Hepatocyte growth factor (HGF) improves alveolar epithelial wound repair in the lung. Stem cells were shown to play a major role in lung injury, repair and fibrosis. We studied the presence, origin and antifibrotic properties of HGF-expressing stem cells in usual interstitial pneumonia. Methods Immunohistochemistry was performed in lung tissue sections and primary alveolar epithelial cells obtained from patients with usual interstitial pneumonia (UIP, n = 7). Bone marrow derived stromal cells (BMSC) from adult male rats were transfected with HGF, instilled intratracheally into bleomycin injured rat lungs and analyzed 7 and 14 days later. Results In UIP, HGF was expressed in specific cells mainly located in fibrotic areas close to the hyperplastic alveolar epithelium. HGF-positive cells showed strong co-staining for the mesenchymal stem cell markers CD44, CD29, CD105 and CD90, indicating stem cell origin. HGF-positive cells also co-stained for CXCR4 (HGF+/CXCR4+) indicating that they originate from the bone marrow. The stem cell characteristics were confirmed in HGF secreting cells isolated from UIP lung biopsies. In vivo experiments showed that HGF-expressing BMSC attenuated bleomycin induced pulmonary fibrosis in the rat, indicating a beneficial role of bone marrow derived, HGF secreting stem cells in lung fibrosis. Conclusions HGF-positive stem cells are present in human fibrotic lung tissue (UIP) and originate from the bone marrow. Since HGF-transfected BMSC reduce bleomycin induced lung fibrosis in the bleomycin lung injury and fibrosis model, we assume that HGF-expressing, bone-marrow derived stem cells in UIP have antifibrotic properties.


Respiratory Research | 2010

DMF inhibits PDGF-BB induced airway smooth muscle cell proliferation through induction of heme-oxygenase-1

Petra Seidel; Stephanie Goulet; Katrin Hostettler; Michael Tamm; Michael Roth

BackgroundAirway wall remodelling is an important pathology of asthma. Growth factor induced airway smooth muscle cell (ASMC) proliferation is thought to be the major cause of airway wall thickening in asthma. Earlier we reported that Dimethylfumarate (DMF) inhibits platelet-derived growth factor (PDGF)-BB induced mitogen and stress activated kinase (MSK)-1 and CREB activity as well as IL-6 secretion by ASMC. In addition, DMF altered intracellular glutathione levels and thereby reduced proliferation of other cell types.MethodsWe investigated the effect of DMF on PDGF-BB induced ASMC proliferation, on mitogen activated protein kinase (MAPK) activation; and on heme oxygenase (HO)-1 expression. ASMC were pre-incubated for 1 hour with DMF and/or glutathione ethylester (GSH-OEt), SB203580, hemin, cobalt-protoporphyrin (CoPP), or siRNA specific to HO-1 before stimulation with PDGF-BB (10 ng/ml).ResultsPDGF-BB induced ASMC proliferation was inhibited in a dose-dependant manner by DMF. PDGF-BB induced the phosphorylation of ERK1/2 and p38 MAPK, but not of JNK. DMF enhanced the PDGF-BB induced phosphorylation of p38 MAPK and there by up-regulated the expression of HO-1. HO-1 induction inhibited the proliferative effect of PDGF-BB. HO-1 expression was reversed by GSH-OEt, or p38 MAPK inhibition, or HO-1 siRNA, which all reversed the anti-proliferative effect of DMF.ConclusionOur data indicate that DMF inhibits ASMC proliferation by reducing the intracellular GSH level with subsequent activation of p38 MAPK and induction of HO-1. Thus, DMF might reduce ASMC and airway remodelling processes in asthma.


American Journal of Respiratory Cell and Molecular Biology | 2012

Steroids and β2-Agonists Regulate Hyaluronan Metabolism in Asthmatic Airway Smooth Muscle Cells

Eleni Papakonstantinou; Ioannis Klagas; George Karakiulakis; Katrin Hostettler; Chong Teck S’ng; Vassiliki Kotoula; Spasenija Savic; Michael Tamm; Michael Roth

Glycosaminoglycans (GAGs), especially hyaluronic acid (HA), regulate tissue flexibility, cell motility, and inflammation. Airway smooth muscle cells (ASMCs) of patients with asthma exhibit abnormal HA metabolism, which contributes to inflammation and remodeling. Here, we investigated the effects of glucocorticoids and long-acting β(2)-agonists (LABAs) on GAG synthesis and HA metabolism by human primary ASMCs. ASMCs were isolated from airway specimens of 10 patients without asthma and 11 patients with asthma. ASMCs were incubated with glucocorticoids, LABAs, or their combination, as well as with their specific receptor antagonists. Secreted and deposited total GAGs were measured by [(3)H]-glucosamine incorporation. The expression of specific GAGs was determined by ELISA and electrophoresis. The expression of HA synthases (HAS), of hyaluronidases (HYALs), and of the HA receptor CD44 was determined by RT-PCR, immunoblotting in cell cultures, and immunohistochemistry in tissue sections of asthmatic lungs. In serum-activated asthmatic ASMCs, glucocorticoids and LABAs significantly inhibited the increased secretion and deposition of total GAGs, but they stimulated secreted and deposited HA of high molecular mass. This effect was attributed to increased mRNA and protein expression of HAS-1 and to the reduced expression of HYAL-1. Furthermore, drug treatment stimulated the expression of CD44 receptors in asthmatic ASMCs. These effects of the drugs were eliminated by their respective receptor inhibitors. Our findings indicate that the combination of glucocorticoids with LABAs counteracts the pathologic degradation of HA, and thereby may reduce the proinflammatory potential of asthmatic ASMCs.


FEBS Letters | 2011

Tenascin-C triggers fibrin accumulation by downregulation of tissue plasminogen activator

Florence Brellier; Katrin Hostettler; Hans-Rudolf Hotz; Ceyda Ozcakir; Sedat Çöloğlu; Dieudonnée Togbe; Bernard Ryffel; Michael Roth; Ruth Chiquet-Ehrismann

We explored novel functions of tenascin‐C by comparing mouse embryonic fibroblasts (MEFs) proficient or deficient in tenascin‐C expression. Transcript profiling analysis identified tissue plasminogen activator (tPA) as the most consistently over‐expressed gene in all tenascin‐C deficient MEFs. This was confirmed by real‐time PCR as well as by protein expression analysis. In agreement with these observations, tenascin‐C deficient MEFs had an increased capacity to digest fibrin in situ. Consistently, tenascin‐C expression in vivo was found to correlate with fibrin deposition in several diseases associated with tenascin‐C overexpression such as fibrosis, asthma and cancer. In conclusion, the present study suggests a new role of tenascin‐C as a regulator of the fibrinolytic system.


Scientific Reports | 2017

Hepatocyte growth factor secreted by bone marrow stem cell reduce ER stress and improves repair in alveolar epithelial II cells.

Izabela Nita; Katrin Hostettler; Luca Giuseppe Athos Tamò; Michaela Medová; Giuseppe Bombaci; Jun Zhong; Ramanjaneyulu Allam; Yitzhak Zimmer; Michael Roth; Thomas Geiser; Amiq Gazdhar

Idiopathic Pulmonary Fibrosis (IPF) is a progressive, irreversible lung disease with complex pathophysiology. Evidence of endoplasmic reticulum (ER) stress has been reported in alveolar epithelial cells (AEC) in IPF patients. Secreted mediators from bone marrow stem cells (BMSC-cm) have regenerative properties. In this study we investigate the beneficial effects of BMSC-cm on ER stress response in primary AEC and ER stressed A549 cells. We hypothesize that BMSC-cm reduces ER stress. Primary AEC isolated from IPF patients were treated with BMSC-cm. To induce ER stress A549 cells were incubated with Tunicamycin or Thapsigargin and treated with BMSC-cm, or control media. Primary IPF-AEC had high Grp78 and CHOP gene expression, which was lowered after BMSC-cm treatment. Similar results were observed in ER stressed A549 cells. Alveolar epithelial repair increased in presence of BMSC-cm in ER stressed A549 cells. Hepatocyte growth factor (HGF) was detected in biologically relevant levels in BMSC-cm. Neutralization of HGF in BMSC-cm attenuated the beneficial effects of BMSC-cm including synthesis of surfactant protein C (SP-C) in primary AEC, indicating a crucial role of HGF in ER homeostasis and alveolar epithelial repair. Our data suggest that BMSC-cm may be a potential therapeutic option for treating pulmonary fibrosis.


Respiration | 2017

Idiopathic Pulmonary Fibrosis in Switzerland: Diagnosis and Treatment

Manuela Funke-Chambour; Andrea Azzola; Dan Adler; Constance Barazzone-Argiroffo; Christian Benden; Annette Boehler; Pierre-Olivier Bridevaux; Martin Brutsche; Christian F. Clarenbach; Katrin Hostettler; Rebekka Kleiner-Finger; Laurent P. Nicod; Paola M. Soccal; Michael Tamm; Thomas Geiser; Romain Lazor

Idiopathic pulmonary fibrosis (IPF) is a severe progressive and irreversible lung disease. Novel antifibrotic drugs that slow disease progression are now available. However, many issues regarding patient management remain unanswered, such as the choice between available drugs, their use in particular subgroups and clinical situations, time of treatment onset, termination, combination or switch, or nonpharmacologic management. To guide Swiss respiratory physicians in this evolving field still characterized by numerous areas of uncertainty, the Swiss Working Group for interstitial and rare lung diseases of the Swiss Respiratory Society provides a position paper on the diagnosis and treatment of IPF.

Collaboration


Dive into the Katrin Hostettler's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Didier Lardinois

University Hospital of Basel

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eleni Papakonstantinou

Aristotle University of Thessaloniki

View shared research outputs
Top Co-Authors

Avatar

Jun Zhong

Woolcock Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ioannis Klagas

Aristotle University of Thessaloniki

View shared research outputs
Researchain Logo
Decentralizing Knowledge