Network


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

Hotspot


Dive into the research topics where Eva Verena Tretter is active.

Publication


Featured researches published by Eva Verena Tretter.


Acta Anaesthesiologica Scandinavica | 2016

Cyclic and constant hyperoxia cause inflammation, apoptosis and cell death in human umbilical vein endothelial cells.

Jing Wu; Christina Hafner; J. P. Schramel; Christoph Kaun; Konstantin A. Krychtiuk; Johann Wojta; Stefan Boehme; Roman Ullrich; Eva Verena Tretter; Klaus Markstaller; Klaus Ulrich Klein

Perioperative high‐dose oxygen (O2) exposure can cause hyperoxia. While the effect of constant hyperoxia on the vascular endothelium has been investigated to some extent, the impact of cyclic hyperoxia largely remains unknown. We hypothesized that cyclic hyperoxia would induce more injury than constant hyperoxia to human umbilical vein endothelial cells (HUVECs).


Shock | 2017

Hyperoxia Induces Inflammation and Cytotoxicity in Human Adult Cardiac Myocytes.

Christina Hafner; Jing Wu; Akos Tiboldi; Moritz Hess; Goran Mitulovic; Christoph Kaun; Konstantin A. Krychtiuk; Johann Wojta; Roman Ullrich; Eva Verena Tretter; Klaus Markstaller; Klaus Ulrich Klein

ABSTRACT Supplemental oxygen (O2) is used as adjunct therapy in anesthesia, emergency, and intensive care medicine. We hypothesized that excessive O2 levels (hyperoxia) can directly injure human adult cardiac myocytes (HACMs). HACMs obtained from the explanted hearts of transplantation patients were exposed to constant hyperoxia (95% O2), intermittent hyperoxia (alternating 10 min exposures to 5% and 95% O2), constant normoxia (21% O2), or constant mild hypoxia (5% O2) using a bioreactor. Changes in cell morphology, viability as assessed by lactate dehydrogenase (LDH) release and trypan blue (TB) staining, and secretion of vascular endothelial growth factor (VEGF), macrophage migration inhibitory factor (MIF), and various pro-inflammatory cytokines (interleukin, IL; chemokine C-X-C motif ligand, CXC; granulocyte-colony stimulating factor, G-CSF; intercellular adhesion molecule, ICAM; chemokine C-C motif ligand, CCL) were compared among treatment groups at baseline (0 h) and after 8, 24, and 72 h of treatment. Changes in HACM protein expression were determined by quantitative proteomic analysis after 48 h of exposure. Compared with constant normoxia and mild hypoxia, constant hyperoxia resulted in a higher TB-positive cell count, greater release of LDH, and elevated secretion of VEGF, MIF, IL-1&bgr;, IL-6, IL-8, CXCL-1, CXCL-10, G-CSF, ICAM-1, CCL-3, and CCL-5. Cellular inflammation and cytotoxicity gradually increased and was highest after 72 h of constant and intermittent hyperoxia. Quantitative proteomic analysis revealed that hypoxic and hyperoxic O2 exposure differently altered the expression levels of proteins involved in cell-cycle regulation, energy metabolism, and cell signaling. In conclusion, constant and intermittent hyperoxia induced inflammation and cytotoxicity in HACMs. Cell injury occurred earliest and was greatest after constant hyperoxia, but even relatively brief repeating hyperoxic episodes induced a substantial inflammatory response.


European Journal of Cardio-Thoracic Surgery | 2018

Argon preconditioning enhances postischaemic cardiac functional recovery following cardioplegic arrest and global cold ischaemia

Attila Kiss; Huaqing Shu; Ouafa Hamza; David Santer; Eva Verena Tretter; Shanglong Yao; Klaus Markstaller; Seth Hallström; Bruno K. Podesser; Klaus Ulrich Klein

OBJECTIVES Previous studies demonstrated that preconditioning with argon gas provided a marked reduction in inflammation and apoptosis and increased myocardial contractility in the setting of acute myocardial ischaemia-reperfusion (IR). There is substantial evidence that myocardial IR injury following cardioplegic arrest is associated with the enhancement of apoptosis and inflammation, which is considered to play a role in cardiac functional impairment. Therefore, the present study was designed to clarify whether preconditioning with argon gas enhances recovery of cardiac function following cardioplegic arrest. METHODS Sprague-Dawley rats were anaesthetized and ventilated and allocated to (i) the control group (control IR, n = 10) and (ii) the in vivo group (argon IR), which received 3 cycles of argon (50% argon, 21% oxygen and 29% nitrogen, n = 10) administered for 5 min interspersed with 5 min of a gas mixture (79% nitrogen and 21% oxygen). The hearts were excised and then evaluated in an erythrocyte-perfused isolated working heart system. Cold ischaemia (4°C) for 60 min was induced by histidine-tryptophan-ketoglutarate cardioplegia, followed by 40 min of reperfusion. Cardiac functional parameters were assessed. In left ventricular tissue samples, the expressions of extracellular-regulated kinase (ERK1/2), AKT serine/threonine kinase (Akt), jun N-terminal kinase (JNK), endothelial nitric oxide synthase (eNOS) and HMGB1: high-mobility group box 1 (HMGB1) protein were assessed by western blot, and high-energy phosphates were evaluated by high-performance liquid chromatography. RESULTS At the end of reperfusion, the rats preconditioned with argon showed significantly enhanced recovery of cardiac output (101 ± 6% vs 87 ± 11%; P < 0.01), stroke volume (94 ± 4% vs 80 ± 11%; P = 0.001), external heart work (100 ± 6% vs 81 ± 13%; P < 0.001) and coronary flow (90 ± 13% vs 125 ± 21%; P < 0.01) compared with the control IR group. These results were accompanied by a significant increase in the levels of myocardial phosphocreatine (23.71 ± 2.07 µmol/g protein vs the control IR group, 13.50 ± 4.75; P = 0.001) and maintained adenosine triphosphate levels (13.62 ±1.89 µmol/g protein vs control IR group adenosine triphosphate: 10.08 ± 1.94 µmol/g; P = 0.017). Additionally, preconditioning with argon markedly reduced the activation of JNK (0.11 ± 0.01 vs 0.25 ± 0.03; P = 0.005) and the expression of HMGB1 protein (0.52 ± 0.04 vs 1.5 ± 0.10; P < 0.001) following reperfusion. CONCLUSIONS Preconditioning with argon enhanced cardiac functional recovery in rat hearts arrested with histidine-tryptophan-ketoglutarate cardioplegia, thereby representing a potential novel cardioprotective approach in cardiac surgery.


Biotechnology Journal | 2017

Differences in stem cell processing lead to distinct secretomes secretion — implications for differential results of previous clinical trials of stem cell therapy for myocardial infarction†

Bernhard Wernly; I Goncalves; Attila Kiss; Vera Paar; Tobias Mösenlechner; Michael Leisch; David Santer; Lucas Motloch; Klaus Ulrich Klein; Eva Verena Tretter; Daniel Kretzschmar; Bruno K. Podesser; Christian Jung; Uta C. Hoppe; Michael Lichtenauer

Stem cell therapy for acute myocardial infarction (AMI) seemed to be a promising therapy, however, large clinical trials brought differential outcome. It has been shown that paracrine effects of secretomes of stem cells rather than cell therapy might play a fundamental role. The present study seeks to compare cell processing protocols of clinical trials and investigate effects of differential cell culture conditions on chemokine secretion and functional effects. Different secretomes are compared regarding IL-8, VEGF, MCP-1, and TNF-alpha secretion. Secretome mediated effects are evaluated on endothelial cell (HUVEC) tube formation and migration. Cardioprotective signaling kinases in human cardiomyocytes are determined by Western immunoblotting. Cells processed according to the REPAIR-AMI protocol secrete significantly higher amounts of IL-8 (487.3 ± 1231.1 vs 9.1 ± 8.2 pg mL-1 ; p < 0.05). REAPIR-AMI supernatants lead to significantly pronounced tube formation and migration on HUVEC and enhance the phosphorylation of Akt, ERK, and CREB. Cell processing conditions have a major impact on the composition of the secretome. The REPAIR-AMI secretome significantly enhances proangiogenic chemokine secretion, angiogenesis, cell migration, and cardioprotective signaling pathways. These results might explain differential outcomes between clinical trials. Optimizing cell processing protocols with special regards to paracrine factors, might open a new therapeutic concept for improving patient outcome.


European Surgical Research | 2016

Argon Preconditioning Protects Airway Epithelial Cells against Hydrogen Peroxide-Induced Oxidative Stress

Christina Hafner; Hong Qi; Lourdes Soto-Gonzalez; Katharina Doerr; Roman Ullrich; Eva Verena Tretter; Klaus Markstaller; Klaus Ulrich Klein

Background: Oxidative stress is the predominant pathogenic mechanism of ischaemia-reperfusion (IR) injury. The noble gas argon has been shown to alleviate oxidative stress-related myocardial and cerebral injury. The risk of lung IR injury is increased in some major surgeries, reducing clinical outcome. However, no study has examined the lung-protective efficacy of argon preconditioning. The present study investigated the protective effects of argon preconditioning on airway epithelial cells exposed to hydrogen peroxide (H2O2) to induce oxidative stress. Methods: A549 airway epithelial cells were treated with a cytotoxic concentration of H2O2 after exposure to standard air or 30 or 50% argon/21% oxygen/5% carbon dioxide/rest nitrogen for 30, 45 or 180 min. Cells were stained with annexin V/propidium iodide, and apoptosis was evaluated by fluorescence-activated cell sorting. Protective signalling pathways activated by argon exposure were identified by Western blot analysis for phosphorylated candidate molecules of the mitogen-activated protein kinase and protein kinase B (Akt) pathways. Results: Preconditioning with 50% argon for 30, 45 and 180 min and 30% argon for 180 min caused significant protection of A549 cells against H2O2-induced apoptosis, with increases in cellular viability of 5-47% (p < 0.0001). A small adverse effect was also observed, which presented as a 12-15% increase in cellular necrosis in argon-treated groups. Argon exposure resulted in early activation of c-Jun N-terminal kinase (JNK) and p38, peaking 10- 30 min after the start of preconditioning, and delayed activation of the extracellular signal-regulated kinase 1/2 (ERK1/2) pathway, peaking after 60-90 min. Conclusions: Argon preconditioning protects airway epithelial cells from H2O2-induced apoptotic cell death. Argon activates the JNK, p38, and ERK1/2 pathways, but not the Akt pathway. The cytoprotective properties of argon suggest possible prophylactic applications in surgery-related IR injury of the lungs.


European Surgical Research | 2016

Contents Vol. 57, 2016

Eun Jin Ahn; Hyun Kang; Geun Joo Choi; Chong Wha Baek; Yong Hun Jung; Young Cheol Woo; Si Ra Bang; Tonia Jeiter; Petra Ruemmele; Stefan M. Brunner; Ulrich Hahn; Rebecca Kesselring; Christoph Rubner; Zsolt Sziklavari; Hans S. Hofmann; Hans J. Schlitt; Stefan Fichtner-Feigl; Olaf Dirsch; Uta Dahmen; Franziska Mußbach; Utz Settmacher; Chichi Xie; Sabrina Ehnert; Romina Haydée Aspera-Werz; Thomas Freude; Marie Karolina Reumann; Björn Gunnar Ochs; Christian Bahrs; Steffen Schröter; Elke Wintermeyer

263 20th Surgical Research Days, Section of Surgical Research of the German Society of Surgery September 8–10, 2016, Magdeburg, Germany Guest Editors: Bruns, C. (Cologne); Halangk, W. (Magdeburg) (available online only) 336 ESSR News Suppl. 1 The 51st Congress of the European Society for Surgical Research May 25–28, 2016, Prague, Czech Republic Abstracts Guest Editors: Froněk, J.; Chlupáč, J.; Malý, S.; Pantoflíček, T.; Ryska, M. (Prague) (available online only)


Journal of Hypertension | 2017

Tenascin-C promotes chronic pressure overload-induced cardiac dysfunction, hypertrophy and myocardial fibrosis

Bruno K. Podesser; Maximilian Kreibich; Elda Dzilic; David Santer; Lorenz Förster; Sandra Trojanek; Dietmar Abraham; Martin Krssak; Klaus Ulrich Klein; Eva Verena Tretter; Christoph Kaun; Johann Wojta; Barbara Kapeller; I Goncalves; Karola Trescher; Attila Kiss


Cardiovascular Research | 2018

P261The role of tenascin C under hyperglycaemic and hypertrophic conditions - In vitro H9c2 rat cardiomyoblasts model

I Fonseca Goncalves; E Acar; Eva Verena Tretter; U Klein; Attila Kiss; Bruno K. Podesser


Cardiovascular Research | 2018

P260The role of tenascin C under hypoxic and hypertrophic conditions - in vitro H9c2 rat cardiomyoblasts model and potential miRNA targeting approach

I Fonseca Goncalves; E Acar; Eva Verena Tretter; U Klein; Bruno K. Podesser; Attila Kiss


Thoracic and Cardiovascular Surgeon | 2017

Remote Ischemic Conditioning Improves Post-Ischemic Cardiac Function: The Role of Neuregulin-1

Attila Kiss; P. Pilz; I Goncalves; M. Inci; F. Nagel; Eva Verena Tretter; Klaus Ulrich Klein; Bruno K. Podesser

Collaboration


Dive into the Eva Verena Tretter's collaboration.

Top Co-Authors

Avatar

Klaus Ulrich Klein

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Attila Kiss

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Bruno K. Podesser

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Klaus Markstaller

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Christina Hafner

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Christoph Kaun

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

David Santer

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

I Goncalves

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Johann Wojta

Medical University of Vienna

View shared research outputs
Top Co-Authors

Avatar

Roman Ullrich

Medical University of Vienna

View shared research outputs
Researchain Logo
Decentralizing Knowledge