Network


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

Hotspot


Dive into the research topics where S. Rosseau is active.

Publication


Featured researches published by S. Rosseau.


Respiratory Research | 2006

Streptococcus pneumoniae induced c-Jun-N-terminal kinase- and AP-1 -dependent IL-8 release by lung epithelial BEAS-2B cells

Bernd Schmeck; Kerstin Moog; Janine Zahlten; Vincent van Laak; Philippe Dje N'Guessan; Bastian Opitz; S. Rosseau; Norbert Suttorp; Stefan Hippenstiel

BackgroundAlthough pneumococcal pneumonia is one of the most common causes of death due to infectious diseases, little is known about pneumococci-lung cell interaction. Herein we tested the hypothesis that pneumococci activated pulmonary epithelial cell cytokine release by c-Jun-NH2-terminal kinase (JNK)MethodsHuman bronchial epithelial cells (BEAS-2B) or epithelial HEK293 cells were infected with S. pneumoniae R6x and cytokine induction was measured by RT-PCR, ELISA and Bioplex assay. JNK-phosphorylation was detected by Western blot and nuclear signaling was assessed by electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP). JNK was modulated by the small molecule inhibitor SP600125 and AP1 by transfection of a dominant negative mutant.ResultsS. pneumoniae induced the release of distinct CC and CXC, as well as Th1 and Th2 cytokines and growth factors by human lung epithelial cell line BEAS-2B. Furthermore, pneumococci infection resulted in JNK phosphorylation in BEAS-2B cells. Inhibition of JNK by small molecule inhibitor SP600125 reduced pneumococci-induced IL-8 mRNA expression and release of IL-8 and IL-6. One regulator of the il8 promoter is JNK-phosphorylated activator protein 1 (AP-1). We showed that S. pneumoniae time-dependently induced DNA binding of AP-1 and its phosphorylated subunit c-Jun with a maximum at 3 to 5 h after infection. Recruitment of Ser63/73-phosphorylated c-Jun and RNA polymerase II to the endogenous il8 promoter was found 2 h after S. pneumoniae infection by chromatin immunoprecipitation. AP-1 repressor A-Fos reduced IL-8 release by TLR2-overexpressing HEK293 cells induced by pneumococci but not by TNFα. Antisense-constructs targeting the AP-1 subunits Fra1 and Fra2 had no inhibitory effect on pneumococci-induced IL-8 release.ConclusionS. pneumoniae-induced IL-8 expression by human epithelial BEAS-2B cells depended on activation of JNK and recruitment of phosphorylated c-Jun to the il8 promoter.


Thorax | 2010

Adrenomedullin attenuates ventilator-induced lung injury in mice

Holger C. Müller; Martin Witzenrath; Thomas Tschernig; Birgitt Gutbier; Stefan Hippenstiel; Ansgar Santel; Norbert Suttorp; S. Rosseau

Background Mechanical ventilation (MV) is a life-saving intervention in acute respiratory failure without any alternative. However, even protective ventilation strategies applying minimal mechanical stress may evoke ventilator-induced lung injury (VILI). Adjuvant pharmacological strategies in addition to lung-protective ventilation to attenuate VILI are lacking. Adrenomedullin exhibited endothelial barrier-stabilising properties in vitro and in vivo. Methods In untreated mice (female C57/Bl6 mice, 11–15 weeks old) and animals treated with adrenomedullin, lung permeability, local and systemic inflammation and markers of distal organ function were assessed following 2 or 6 h of mechanical ventilation with 100% oxygen and protective or moderately injurious ventilator settings, respectively. Results Adrenomedullin dramatically reduced lung permeability in VILI in mice, leading to improved oxygenation. Adrenomedullin treatment reduced myosin light chain phosphorylation, attenuated the accumulation of leucocytes in the lung and prevented the increase in lactate and creatinine levels in mice ventilated with high tidal volumes. Moreover, adrenomedullin protected against VILI even when treatment was initiated 2 h after the beginning of mechanical ventilation in a 6 h VILI mouse model. Conclusion Adjuvant treatment with adrenomedullin may be a promising new pharmacological approach to attenuate VILI.


Immunology | 2007

Comparative transcriptional profiling of the lung reveals shared and distinct features of Streptococcus pneumoniae and influenza A virus infection.

S. Rosseau; Andreas C. Hocke; Hans J. Mollenkopf; Bernd Schmeck; Norbert Suttorp; Stefan H. E. Kaufmann; Jens Zerrahn

Pneumonia is the most common cause of death from infectious disease in the western hemisphere. Pathophysiological and protective processes are initiated by pattern recognition of microbial structures. To provide the molecular framework for a better understanding of processes relevant to host defence in pneumonia, we performed pulmonary transcriptome analysis in mice infected with the major bacterial and viral agents of community‐acquired pneumonia, Streptococcus pneumoniae and influenza A virus. We detected differential expression of 1300 genes after infection with either pathogen. Of these, approximately 36% or 30% were specific for pneumococcal or influenza infection, respectively, yielding pathogen‐specific as well as shared inflammatory transcriptional signatures. These results not only reveal a differential response on the cytokine and chemokine levels but also emphasize the important role of genes implicated in regulation and fine tuning of inflammation. As one, albeit unexpected, key feature of pneumococcal pneumonia we discovered down‐regulation of B‐cell responses, probably reflecting a pneumococcal virulence strategy. The pathophysiological consequences of influenza A virus infection were reflected by the emerging protective T‐cell response and differential induction of genes involved in tissue regeneration and proliferation. These data provide new insights into pathogenesis of the most common forms of pneumonia, highlighting the value of transcriptional profiling for the elucidation of underlying mechanisms.


Pulmonary Pharmacology & Therapeutics | 2011

The Sphingosine-1 Phosphate receptor agonist FTY720 dose dependently affected endothelial integrity in vitro and aggravated ventilator-induced lung injury in mice.

Holger C. Müller; Andreas C. Hocke; K Hellwig; Birgitt Gutbier; Harm Peters; Stefanie M. Schönrock; Thomas Tschernig; Andreas Schmiedl; Stefan Hippenstiel; Philippe Dje N’Guessan; S. Rosseau; Norbert Suttorp; Martin Witzenrath

Lung barrier protection by Sphingosine-1 Phosphate (S1P) has been demonstrated experimentally, but recent evidence suggests barrier disruptive properties of high systemic S1P concentrations. The S1P analog FTY720 recently gained an FDA approval for treatment of multiple sclerosis. In case of FTY720 treated patients experiencing multiple organ dysfunction syndrome the drug may accumulate due to liver failure, and the patients may receive ventilator therapy. Whereas low doses of FTY720 enhanced endothelial barrier function, data on effects of increased FTY720 concentrations are lacking. We measured transcellular electrical resistance (TER) of human umbilical vein endothelial cell (HUVEC) monolayers, performed morphologic analysis and measured apoptosis by TUNEL staining and procaspase-3 degradation in HUVECs stimulated with FTY720 (0.01-100 μM). Healthy C57BL/6 mice and mice ventilated with 17 ml/kg tidal volume and 100% oxygen for 2 h were treated with 0.1 or 2 mg/kg FTY720 or solvent, and lung permeability, oxygenation and leukocyte counts in BAL and blood were quantified. Further, electron microscopic analysis of lung tissue was performed. We observed barrier protective effects of FTY720 on HUVEC cell layers at concentrations up to 1 μM while higher concentrations induced irreversible barrier breakdown accompanied by induction of apoptosis. Low FTY720 concentrations (0.1 mg/kg) reduced lung permeability in mechanically ventilated mice, but 2 mg/kg FTY720 increased pulmonary vascular permeability in ventilated mice accompanied by endothelial apoptosis, while not affecting permeability in non-ventilated mice. Moreover, hyperoxic mechanical ventilation sensitized the pulmonary vasculature to a barrier disrupting effect of FTY720, resulting in worsening of ventilator induced lung injury. In conclusion, the current data suggest FTY720 induced endothelial barrier dysfunction, which was probably caused by proapoptotic effects and enhanced by mechanical ventilation.


Deutsches Arzteblatt International | 2013

Extracorporeal Lung Support in H1N1 Provoked Acute Respiratory Failure: The Experience of the German ARDS Network

Steffen Weber-Carstens; Anton Goldmann; Michael Quintel; Armin Kalenka; Stefan Kluge; Jürgen Peters; Christian Putensen; Thomas Müller; S. Rosseau; Bernhard Zwißler; Onnen Moerer

BACKGROUND During the H1N1 pandemic of 2009 and 2010, the large number of patients with severe respiratory failure due to H1N1 infection strained the capacities of treatment facilities for extracorporeal membrane oxygenation (ECMO) around the world. No data on this topic have yet been published for Germany. METHODS During the pandemic, the German ARDS Network (a task force of the DIVIs respiratory failure section) kept track of the availability of ECMO treatment facilities with a day-to-day, Internet-based capacity assessment. In cooperation with the Robert Koch Institute, epidemiological and clinical data were obtained on all patients treated for influenza in intensive care units. RESULTS 116 patients were identified who had H1N1 disease and were treated in the intensive care units of 9 university hospitals and 3 other maximum medical care hospitals. 61 of them received ECMO. The overall mortality was 38% (44 of 116 patients); among patients receiving ECMO, the mortality was 54% (33 of 61 patients). The mortality was higher among patients who had an accompanying malignancy or immune deficiency (72.2%). CONCLUSION Even persons without any other accompanying disease developed life-threatening respiratory failure as a result of H1N1 infection, and many of these patients needed ECMO. This study reveals for the first time that the mortality of H1N1 infection in Germany is comparable to that in other countries. H1N1 patients with acute respiratory failure had a worse outcome if they also had serious accompanying diseases.


Experimental and Toxicologic Pathology | 2008

Rho-kinase and contractile apparatus proteins in murine airway hyperresponsiveness

Martin Witzenrath; B. Ahrens; Bernd Schmeck; Stefanie M. Kube; Stefan Hippenstiel; S. Rosseau; Eckard Hamelmann; Norbert Suttorp; Hartwig Schütte

Airway hyperresponsiveness (AHR) is a hallmark of bronchial asthma. Increased expression of smooth muscle contractile proteins or increased responsiveness of the contractile apparatus due to RhoA/Rho-kinase activation may contribute to AHR. BALB/c mice developed AHR following systemic sensitization by intraperitoneal injections of 20 microg ovalbumin (OVA) in presence of 2mg Al(OH)(3) on days 1 and 14, and airway challenge by 1% OVA-inhalation for 20 min each on days 28, 29 and 30. As assessed by Western blot, protein expression of RhoA, MLC (myosin light chain) and smMLCK (smooth muscle myosin light chain kinase) was increased in lungs of OVA/OVA-animals with AHR, as well as in lungs of OVA-sensitized and sham-challenged animals (OVA/PBS) without AHR, compared with lungs of PBS/PBS-animals. Pretreatment with the specific Rho-kinase inhibitor Y-27632 reduced MLC-phosphorylation and AHR. Contribution of Rho-kinase to bronchoconstriction was increased in lungs of OVA/OVA-animals compared with OVA/PBS- and PBS/PBS-animals, respectively. Furthermore, bronchoconstriction following MCh stimulation was significantly reduced after Y-27632 application. In conclusion, systemic allergen-sensitization increased pulmonary expression of proteins involved in smooth muscle contraction, which may contribute to development of AHR. However, this observation was independent from local allergen challenge, suggesting that additional cofactors may be required for the activation of Rho-kinase and thereby the induction of AHR. Rho-kinase may play an important role in murine AHR, and the bronchodilating action of Rho-kinase inhibition may offer a new therapeutic perspective in obstructive airway disease.


Amyotrophic Lateral Sclerosis | 2017

Clinical characteristics and course of dying in patients with amyotrophic lateral sclerosis withdrawing from long-term ventilation.

Dagmar Kettemann; Andreas Funke; André Maier; S. Rosseau; Robert Meyer; Susanne Spittel; C. Münch; Thomas Meyer

Abstract Non-invasive ventilation (NIV) or tracheotomy with invasive ventilation (TIV) are treatment options in ALS. However, a proportion of patients receiving long-term ventilation decide to have it withdrawn. The objective of this study was to analyse the clinical characteristics and palliative approaches in ALS patients withdrawing from long-term ventilation (WLTV). In a cohort study, two different palliative concepts in WLTV were studied: (1) augmented symptom control (ASC; sedation not intended) in patients with ventilator-free tolerance; (2) continuous deep sedation (CDS; sedation intended) in patients without ventilator-free tolerance. Results showed that WLTV was realised in 49 ALS patients (NIV = 13; TIV = 36). Mean daily ventilation was 23.4 h. The ALS Functional Rating Scale (ALSFRS-R) was low (5.6 of 48). Forty-one per cent of patients (n = 20) presented with ophthalmoplegia. ASC was performed in 20 patients, CDS in 29 patients. The mean time to death following disconnection was 32 (0.3–164) h during ASC and 0.3 (0.2–0.6) h in CDS. In conclusion, a low ALSFRS-R, high incidence of ophthalmoplegia and extended ventilator dependency were found before WLTV. The presence or absence of ventilator-free tolerance determined the approach to the management of symptoms, the setting for immediate end-of-life care and the course of dying in WLTV.


Critical Care | 2017

Outcome of acute respiratory distress syndrome in university and non-university hospitals in Germany

Konstantinos Raymondos; Tamme Dirks; Michael Quintel; Ulrich Molitoris; Jörg Ahrens; Thorben Dieck; Kai Johanning; Dietrich Henzler; Rolf Rossaint; Christian Putensen; Hermann Wrigge; Ralph Wittich; M. Ragaller; Thomas Bein; Martin Beiderlinden; Maxi Sanmann; Christian Rabe; Jörn Schlechtweg; Monika Holler; Fernando Frutos-Vivar; Andrés Esteban; Hartmut Hecker; S. Rosseau; Vera von Dossow; Claudia Spies; Tobias Welte; S. Piepenbrock; Steffen Weber-Carstens

BackgroundThis study investigates differences in treatment and outcome of ventilated patients with acute respiratory distress syndrome (ARDS) between university and non-university hospitals in Germany.MethodsThis subanalysis of a prospective, observational cohort study was performed to identify independent risk factors for mortality by examining: baseline factors, ventilator settings (e.g., driving pressure), complications, and care settings—for example, case volume of ventilated patients, size/type of intensive care unit (ICU), and type of hospital (university/non-university hospital). To control for potentially confounding factors at ARDS onset and to verify differences in mortality, ARDS patients in university vs non-university hospitals were compared using additional multivariable analysis.ResultsOf the 7540 patients admitted to 95 ICUs from 18 university and 62 non-university hospitals in May 2004, 1028 received mechanical ventilation and 198 developed ARDS. Although the characteristics of ARDS patients were very similar, hospital mortality was considerably lower in university compared with non-university hospitals (39.3% vs 57.5%; p = 0.012). Treatment in non-university hospitals was independently associated with increased mortality (OR (95% CI): 2.89 (1.31–6.38); p = 0.008). This was confirmed by additional independent comparisons between the two patient groups when controlling for confounding factors at ARDS onset. Higher driving pressures (OR 1.10; 1 cmH2O increments) were also independently associated with higher mortality. Compared with non-university hospitals, higher positive end-expiratory pressure (PEEP) (mean ± SD: 11.7 ± 4.7 vs 9.7 ± 3.7 cmH2O; p = 0.005) and lower driving pressures (15.1 ± 4.4 vs 17.0 ± 5.0 cmH2O; p = 0.02) were applied during therapeutic ventilation in university hospitals, and ventilation lasted twice as long (median (IQR): 16 (9–29) vs 8 (3–16) days; p < 0.001).ConclusionsMortality risk of ARDS patients was considerably higher in non-university compared with university hospitals. Differences in ventilatory care between hospitals might explain this finding and may at least partially imply regionalization of care and the export of ventilatory strategies to non-university hospitals.


Pneumologie | 2010

Management der Influenza A/H1N1 – Pandemie im Krankenhaus: Update Januar 2010

Tom Schaberg; Tt Bauer; Klaus Dalhoff; Santiago Ewig; D. Köhler; J. Lorenz; Gernot Rohde; M. W. Pletz; S. Rosseau; B. Schaaf; Norbert Suttorp; T. Welte; W. Haas; A. Reuß

This update summarized the hospital experience from the first wave of the new influenza A/H1/N1 pandemic.


Der Internist | 2007

New aspects of the pathophysiology of pneumonia

Stefan Hippenstiel; Martin Witzenrath; Bastian Opitz; Hartwig Schütte; S. Rosseau; Norbert Suttorp

ZusammenfassungPneumonien können zu vital bedrohlichen Störungen der Gasaustauschfunktion der Lunge führen. Bedingt durch die große Vielfalt der Pneumonieerreger wirken unterschiedliche Virulenzfaktoren auf die Lunge ein. Es ist von besonderer Bedeutung, dass neben den direkt pathogenassoziierten Schädigungen eine überschießende Entzündungsreaktion zu Versagen von pulmonaler Barriere und Gasaustauschfunktion führen kann. Neben unspezifischen Abwehrmechanismen kommt rezeptorvermittelter Aktivierung zellgebundener angeborener Immunität eine zentrale Rolle für die pulmonale Immunantwort zu. Pathogenassoziierte Moleküle werden durch transmembranäre und zytosolische Rezeptoren des Wirtes erkannt. Diese Interaktion führt über Signalkaskaden zur Expression immunmodulatorischer Moleküle. Über die adäquate Antibiotikatherapie hinaus müssen innovative Therapieansätze zur Pneumoniebehandlung überschießende Entzündungsreaktionen begrenzen unter gleichzeitiger Unterstützung pulmonaler Abwehrkapazität.AbstractPneumonia can lead to the critical impairment of gas exchange in the lung. Due to the great variability of pneumonia causing pathogens, a large variety of diverse virulence factors act on the lung. Besides stimulation of unspecific defense mechanisms, activation of receptor-dependent cell-mediated innate immune defense mechanisms are critical for the pulmonary immune defense. Pathogen-associated molecules are detected via transmembraneous and cytosolic receptors of the host. This interaction stimulates the expression of immunomodulatory molecules via signal cascades. Of particular importance, in addition to direct pathogen-caused lung damage, is the overwhelming activation of the inflammatory response which can result in lung barrier failure and impairment of pulmonary gas exchange. In addition to the design of new antibiotics, innovative therapeutic strategies should therefore concentrate on the enhancement of antimicrobial mechanisms by concurrent limitation of inflammation.

Collaboration


Dive into the S. Rosseau's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

N Suttorp

Humboldt State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge