Network


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

Hotspot


Dive into the research topics where Iris Müller is active.

Publication


Featured researches published by Iris Müller.


Journal of Experimental Medicine | 2002

A Critical Role of Platelet Adhesion in the Initiation of Atherosclerotic Lesion Formation

Steffen Massberg; Korbinian Brand; Sabine Grüner; Sharon Page; Elke Müller; Iris Müller; Wolfgang Bergmeier; Thomas Richter; Michael Lorenz; Ildiko Konrad; Bernhard Nieswandt; Meinrad Gawaz

The contribution of platelets to the process of atherosclerosis remains unclear. Here, we show in vivo that platelets adhere to the vascular endothelium of the carotid artery in ApoE − / − mice before the development of manifest atherosclerotic lesions. Platelet–endothelial cell interaction involved both platelet glycoprotein (GP)Ibα and GPIIb-IIIa. Platelet adhesion to the endothelium coincides with inflammatory gene expression and preceded atherosclerotic plaque invasion by leukocytes. Prolonged blockade of platelet adhesion in ApoE − / − mice profoundly reduced leukocyte accumulation in the arterial intima and attenuated atherosclerotic lesion formation in the carotid artery bifurcation, the aortic sinus, and the coronary arteries. These findings establish the platelet as a major player in initiation of the atherogenetic process.


Heart | 2001

Effect of a high loading dose of clopidogrel on platelet function in patients undergoing coronary stent placement

Iris Müller; Melchior Seyfarth; Silja Rüdiger; Beate Wolf; Gisela Pogatsa-Murray; Albert Schömig; Meinrad Gawaz

Following coronary stent placement, platelet activation is a major determinant of the risk of subacute stent thrombosis.1 Combined antiplatelet treatment with ticlopidine and aspirin reduced platelet activation after coronary stenting1. Although combined antiplatelet treatment consisting of aspirin and ticlopidine has significantly reduced early ischaemic events following coronary stenting, stent thrombosis still occurs in up to 1% of treated patients, especially in the early days after the intervention, probably because of delayed onset of action of ticlopidine. Clopidogrel is a ticlopidine-like novel thienopyridine inhibitor of ADP induced platelet activation.2 Clopidogrel differs from ticlopidine in that it has a favourable safety profile compared to ticlopidine and reveals an accelerated antiplatelet activity after first administration. The present study sought to investigate the antiplatelet effect of various doses of clopidogrel in patients undergoing coronary stent placement; comparison was made with standard ticlopidine treatment. Thirty patients were randomised into three treatment arms: group I (n = 10), ticlopidine 2 × 500 g as loading dose and 2 × 250 mg daily thereafter; group II (n = 10), clopidogrel 1 × 300 mg loading dose and 1 × 75 mg per day; or group III (n = 10), clopidogrel 1 × 600 mg plus 2 × 75 mg daily thereafter. All patients received aspirin 2 × 100 mg per day concomitantly. Peripheral venous blood samples were taken with a loose tourniquet through a short venous catheter inserted into a forearm vein before and then 2, …


The FASEB Journal | 2003

Soluble glycoprotein VI dimer inhibits platelet adhesion and aggregation to the injured vessel wall in vivo

Steffen Massberg; Ildiko Konrad; Andreas Bültmann; Christian Schulz; Götz Münch; Mario Peluso; Michael Lorenz; Simon Schneider; Felicitas Besta; Iris Müller; Bin Hu; Harald Langer; Elisabeth Kremmer; Martina Rudelius; Ulrich Heinzmann; Martin Ungerer; Meinrad Gawaz

Platelet—collagen interactions play a fundamental role in the process of arterial thrombosis. The major platelet collagen receptor is the glycoprotein VI (GPVI). Here, we determined the effects of a soluble dimeric form of GPVI on platelet adhesion in vitro and in vivo. We fused the extracellular domain of GPVI with the human immunoglobulin Fc domain. The soluble dimeric form of GPVI (GPVI‐Fc) specifically bound to immobilized collagen. Binding of GPVI‐Fc to collagen was inhibited competitively by soluble GPVI‐Fc, but not control Fc lacking the external GPVI domain. GPVI‐Fc inhibited the adhesion of CHO cells that stably express human GPVI and of platelets on collagen and attenuated thrombus formation under shear conditions in vitro. To test the effects of GPVI‐Fc in vivo, arterial thrombosis was induced in the mouse carotid artery, and platelet—vessel wall interactions were visualized by intravital fluorescence microscopy. Infusion of GPVI‐Fc but not of control Fc virtually abolished stable arrest and aggregation of platelets following vascular injury. Importantly, GPVI‐Fc but not control Fc, was detected at areas of vascular injury. These findings further substantiate the critical role of the collagen receptor GPVI in the initiation of thrombus formation at sites of vascular injury and identify soluble GPVI as a promising antithrombotic strategy.


Circulation | 2003

Effects of Statins on Platelet Inhibition by a High Loading Dose of Clopidogrel

Iris Müller; Felicitas Besta; Christian Schulz; Zhongyan Li; Steffen Massberg; Meinrad Gawaz

Background—Recent studies suggested that some HMG-CoA reductase blockers might inhibit the antiplatelet activity of clopidogrel. Therefore, we analyzed how various statins together with a high loading dose of clopidogrel (600 mg) affect platelet aggregation. Methods and Results—Seventy-seven patients with stable angina scheduled for elective coronary stenting were studied. Patients were randomized to receive atorvastatin, fluvastatin, lovastatin, pravastatin, simvastatin (each 20 mg), cerivastatin (0.3 mg), or placebo, plus a high loading dose of 600 mg of clopidogrel. ADP-induced platelet aggregation (5 and 20 &mgr;mol/L) was determined before and 2 and 4 hours after first clopidogrel administration. All patients were taking aspirin (100 mg/d) regularly. We found that none of the statins significantly influenced inhibition of platelet aggregation by clopidogrel. Conclusions—Concomitant use of statins with clopidogrel does not significantly inhibit antiplatelet activity, at least when clopidogrel is administered at a high loading dose of 600 mg.


Thrombosis Research | 2001

Differential Antiplatelet Effects of Various Glycoprotein IIb-IIIa Antagonists

Timm Dickfeld; Andreas Ruf; Gisela Pogatsa-Murray; Iris Müller; Bernd Engelmann; Winfried Taubitz; Jörg Fischer; Oliver Meier; Meinrad Gawaz

The blockade of platelet glycoprotein IIb-IIIa (GPIIb-IIIa) was recently introduced as a new antiplatelet strategy. At present, various GPIIb-IIIa inhibitors are available to treat patients with acute coronary syndrome or when undergoing percutaneous coronary interventions. The current study systematically evaluates the antiplatelet effects of GPIIb-IIIa inhibitors in clinical use. Using conformation-dependent monoclonal antibodies [ligand-induced binding sites (LIBS-1), PMI-1] and flow cytometry, we showed that the GPIIb-IIIa antagonists abciximab, integrelin, lamifiban, and tirofiban, but not EMD 122347 or YM 337, induced LIBS activity of platelet GPIIb-IIIa. The LIBS activity of GPIIb-IIIa antagonists correlates with a proaggregatory response of fixed platelets pretreated with GPIIb-IIIa antagonists (intrinsic activity). All tested GPIIb-IIIa antagonists completely inhibit concentration-dependent ADP (20 micromol/l)-induced aggregation. In contrast, substantial TRAP (25 micromol/l)-induced platelet aggregation still occurs even at high inhibitor concentrations of the tested GPIIb-IIIa antagonists. In addition, we show that GPIIb-IIIa antagonists are poor inhibitors of platelet release reaction (ATP and P-selectin secretion) especially when strong agonists such as TRAP are used to activate platelets. Inhibition of platelet procoagulant activity (thrombin generation) by GPIIb-IIIa antagonists is dependent on the type and concentration of antagonists and on the strength of stimulus (thrombin, tissue factor) used to induce platelet-dependent thrombin generation. The present data show that significant pharmacological differences exist between GPIIb-IIIa antagonists that may have consequences for antithrombotic strategies and for future drug development.


Pathophysiology of Haemostasis and Thrombosis | 2002

Effects of Aspirin and Clopidogrel versus Oral Anticoagulation on Platelet Function and on Coagulation in Patients with Nonvalvular Atrial Fibrillation (CLAFIB)

Iris Müller; Steffen Massberg; Wolfgang Zierhut; Christiane Binz; Alexander Schuster; Silja Rüdiger-von Hoch; Siegmund Braun; Meinrad Gawaz

The aim of the study was to evaluate the effect of two antithrombotic therapies on platelet function and on coagulation in patients with nonvalvular atrial fibrillation (NVAF). Twenty patients with NVAF were treated with aspirin (300 mg/day) and clopidogrel (75 mg/day) for 2 weeks immediately followed by oral anticoagulation (target international normalized ratio 2.0–3.0). Parameters of platelet function and coagulation were evaluated before antithrombotic therapy, at the end of aspirin plus clopidogrel and during subsequent anticoagulation treatment. Aspirin plus clopidogrel significantly inhibited platelet aggregation, fibrinogen receptor activation and release of P-selectin and prolonged in vitro bleeding time (p < 0.01). Coagulation parameters (platelet-dependent thrombin generation, antithrombin III, thrombin-antithrombin III complex, prothrombin fragment 1 + 2) were not significantly affected. During the subsequent oral anticoagulation phase platelet function was not substantially reduced; however, coagulation parameters were significantly inhibited (p < 0.001). The results indicate that combined antiplatelet therapy is superior to aspirin monotherapy in inhibiting platelet function but does not seem to substantially modulate coagulation cascade in patients with NVAF.


PLOS ONE | 2013

Prognostic value of contrast-enhanced cardiac magnetic resonance imaging in patients with newly diagnosed non-ischemic cardiomyopathy: cohort study.

Karin Müller; Iris Müller; Ulrich Kramer; Reinhard Kandolf; Meinrad Gawaz; Axel Bauer; Christine S. Zuern

Background Owing to its variable course from asymptomatic cases to sudden death risk stratification is of paramount importance in newly diagnosed non-ischemic cardiomyopathy. We tested whether late gadolinium enhancement (LGE) assessed by cardiac magnetic resonance (CMR) imaging is a prognostic marker in consecutive patients with newly diagnosed non-ischemic cardiomyopathy. Methods We enrolled 185 patients who presented for evaluation of newly diagnosed non-ischemic cardiomyopathy. Coronary artery disease was excluded by coronary angiography. Following risk markers were additionally assessed: NYHA functional class (≥II), brain natriuretic peptide (>100 ng/l), troponin I (TnI, ≥0.03 µg/l), left ventricular ejection fraction (LVEF, ≤40%), left ventricular enddiastolic diameter (>55 mm) and QRS duration (>98 ms). Endpoint of the study was the composite of all-cause mortality, heart transplantation, aborted sudden death, sustained ventricular tachycardia or hospitalization due to decompensated heart failure within three years of follow-up. Results During median follow-up of 21 months, 54 patients (29.2%) reached the composite endpoint. Ninety-four of the 185 patients (50.8%) were judged LGE-positive. Prognosis of LGE-positive patients was significantly worse than that of LGE-negative patients (cumulative 3-year event rates of 67.4% in LGE-positive and 27.2% in LGE-negative patients, respectively; p = 0.021). However, in multivariable analysis, presence of LGE was not an independent predictor of outcome. Only LVEF ≤40% and TnI ≥0.03 µg/l were independent risk predictors of the composite endpoint yielding relative risks of 3.9 (95% CI 1.9–8.1; p<0.0001) and 2.2 (95% CI 1.2–4.0; p = 0.014), respectively. Conclusions In consecutive patients presenting with newly diagnosed non-ischemic cardiomyopathy, LGE-positive patients had worse prognosis. However, only traditional risk parameters like left ventricular performance and cardiac biomarkers but not presence of LGE were independent risk predictors.


Journal of Biological Chemistry | 2013

Gremlin-1 is an inhibitor of macrophage migration inhibitory factor and attenuates atherosclerotic plaque growth in ApoE-/- mice

Iris Müller; Tanja Schönberger; Martina Schneider; Oliver Borst; Melanie Ziegler; Peter Seizer; Christoph Leder; Karin Müller; Michael Lang; Florian Appenzeller; Oleg Lunov; Manuela Fahrleitner; Marcus Olbrich; Harald Langer; Tobias Geisler; Florian Lang; Madhumita Chatterjee; Jan de Boer; Uwe J. F. Tietge; Jürgen Bernhagen; Thomas Simmet; Meinrad Gawaz

Results: Gremlin-1 binds with high affinity to macrophage migration inhibitory factor and attenuates the progression of atherosclerosis. Conclusion: We describe a novel mechanism that regulates foam cell formation and plaque growth. Significance: The findings disclose a new mechanism for the regulation of plaque growth and may open novel therapeutic strategies to control the progression of atherosclerosis. Monocyte infiltration and macrophage formation are pivotal steps in atherosclerosis and plaque vulnerability. Gremlin-1/Drm is crucial in embryo-/organogenesis and has been shown to be expressed in the adult organism at sites of arterial injury and to inhibit monocyte migration. The purpose of the present study was to evaluate and characterize the role of Gremlin-1 in atherosclerosis. Here we report that Gremlin-1 is highly expressed primarily by monocytes/macrophages in aortic atherosclerotic lesions of ApoE−/− mice and is secreted from activated monocytes and during macrophage development in vitro. Gremlin-1 reduces macrophage formation by inhibiting macrophage migration inhibitory factor (MIF), a cytokine critically involved in atherosclerotic plaque progression and vulnerability. Gremlin-1 binds with high affinity to MIF (KD = 54 nm), as evidenced by surface plasmon resonance analysis and co-immunoprecipitation, and reduces MIF-induced release of TNF-α from macrophages. Treatment of ApoE−/− mice with a dimeric recombinant fusion protein, mGremlin1-Fc, but not with equimolar control Fc or inactivated mGremlin1-Fc, reduced TNF-α expression, the content of monocytes/macrophages of atherosclerotic lesions, and attenuated atheroprogression. The present data disclose that Gremlin-1 is an endogenous antagonist of MIF and define a role for Gremlin-1/MIF interaction in atherosclerosis.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

Pivotal Role of Serum- and Glucocorticoid-Inducible Kinase 1 in Vascular Inflammation and Atherogenesis

Oliver Borst; Malte Schaub; Britta Walker; Evi Schmid; Patrick Münzer; Jakob Voelkl; Ioana Alesutan; José M. Rodríguez; Sebastian Vogel; Tanja Schoenberger; Katja Metzger; Dominik Rath; Anja T. Umbach; Dietmar Kuhl; Iris Müller; Peter Seizer; Tobias Geisler; Meinrad Gawaz; Florian Lang

Objective— Atherosclerosis, an inflammatory disease of arterial vessel walls, requires migration and matrix metalloproteinase (MMP)-9–dependent invasion of monocytes/macrophages into the vascular wall. MMP-9 expression is stimulated by transcription factor nuclear factor-&kgr;B, which is regulated by inhibitor &kgr;B (I&kgr;B) and thus I&kgr;B kinase. Regulators of nuclear factor-&kgr;B include serum- and glucocorticoid-inducible kinase 1 (SGK1). The present study explored involvement of SGK1 in vascular inflammation and atherogenesis. Approach and Results— Gene-targeted apolipoprotein E (ApoE)–deficient mice without (apoe −/− sgk1 +/+) or with (apoe −/− sgk1 −/−) additional SGK1 knockout received 16-week cholesterol-rich diet. According to immunohistochemistry atherosclerotic lesions in aorta and carotid artery, vascular CD45+ leukocyte infiltration, Mac-3+ macrophage infiltration, vascular smooth muscle cell content, MMP-2, and MMP-9 positive areas in atherosclerotic tissue were significantly less in apoe −/− sgk1 −/−mice than in apoe −/− sgk1 +/+mice. As determined by Boyden chamber, thioglycollate-induced peritonitis and air pouch model, migration of SGK1-deficient CD11b+F4/80+ macrophages was significantly diminished in vitro and in vivo. Zymographic MMP-2 and MMP-9 production, MMP-9 activity and invasion through matrigel in vitro were significantly less in sgk1 −/− than in sgk1 +/+macrophages and in control plasmid–transfected or inactive K127NSGK1-transfected than in constitutively active S422DSGK1-transfected THP-1 cells. Confocal microscopy revealed reduced macrophage number and macrophage MMP-9 content in plaques of apoe −/− sgk1 −/− mice. In THP-1 cells, MMP-inhibitor GM6001 (25 &mgr;mol/L) abrogated S422DSGK1-induced MMP-9 production and invasion. According to reverse transcription polymerase chain reaction, MMP-9 transcript levels were significantly reduced in sgk1 −/−macrophages and strongly upregulated in S422DSGK1-transfected THP-1 cells compared with control plasmid–transfected or K127NSGK1-transfected THP-1 cells. According to immunoblotting and confocal microscopy, phosphorylation of I&kgr;B kinase and inhibitor &kgr;B and nuclear translocation of p50 were significantly lower in sgk1 −/−macrophages than in sgk1 +/+macrophages and significantly higher in S422DSGK1-transfected THP-1 cells than in control plasmid–transfected or K127NSGK1-transfected THP-1 cells. Treatment of S422DSGK1-transfected THP-1 cells with I&kgr;B kinase-inhibitor BMS-345541 (10 &mgr;mol/L) abolished S422DSGK1-induced increase of MMP-9 transcription and gelatinase activity. Conclusions— SGK1 plays a pivotal role in vascular inflammation during atherogenesis. SGK1 participates in the regulation of monocyte/macrophage migration and MMP-9 transcription via regulation of nuclear factor-&kgr;B.


Journal of Biological Chemistry | 2014

Activated Platelets Interfere with Recruitment of Mesenchymal Stem Cells to Apoptotic Cardiac Cells via High Mobility Group Box 1/Toll-like Receptor 4-mediated Down-regulation of Hepatocyte Growth Factor Receptor MET

Sebastian Vogel; Madhumita Chatterjee; Katja Metzger; Oliver Borst; Tobias Geisler; Peter Seizer; Iris Müller; Andreas F. Mack; Susanne Schumann; Hans-Jörg Bühring; Florian Lang; Rüdiger V. Sorg; Harald Langer; Meinrad Gawaz

Background: Mesenchymal stem cells (MSC) contribute to cardiac repair after myocardial injury. Underlying molecular mechanisms remain unexplored. Results: Activated platelets inhibit recruitment of MSC to apoptotic cardiac myocytes and fibroblasts via HMGB1/TLR-4-mediated down-regulation of HGF receptor MET. Conclusion: We identify a novel mechanism by which platelets impair MSC migration to damaged cardiac cells. Significance: The cross-talk between platelets and MSC might be critical for myocardial repair. Recruitment of mesenchymal stem cells (MSC) following cardiac injury, such as myocardial infarction, plays a critical role in tissue repair and may contribute to myocardial recovery. However, the mechanisms that regulate migration of MSC to the site of tissue damage remain elusive. Here, we demonstrate in vitro that activated platelets substantially inhibit recruitment of MSC toward apoptotic cardiac myocytes and fibroblasts. The alarmin high mobility group box 1 (HMGB1) was released by platelets upon activation and mediated inhibition of the cell death-dependent migratory response through Toll-like receptor (TLR)-4 expressed on the MSC. Migration of MSC to apoptotic cardiac myocytes and fibroblasts was driven by hepatocyte growth factor (HGF), and platelet activation was followed by HMGB1/TLR-4-dependent down-regulation of HGF receptor MET on MSC, thereby impairing HGF-driven MSC recruitment. We identify a novel mechanism by which platelets, upon activation, interfere with MSC recruitment to apoptotic cardiac cells, a process that may be of particular relevance for myocardial repair and regeneration.

Collaboration


Dive into the Iris Müller's collaboration.

Top Co-Authors

Avatar

Meinrad Gawaz

Ludwig Maximilian University of Munich

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Seizer

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar

Florian Lang

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dominik Rath

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar

Madhumita Chatterjee

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar

Oliver Borst

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge