Network


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

Hotspot


Dive into the research topics where Katrin Schröder is active.

Publication


Featured researches published by Katrin Schröder.


Hypertension | 2008

Apocynin Is Not an Inhibitor of Vascular NADPH Oxidases but an Antioxidant

Sabine Heumüller; Sven Wind; Eduardo Barbosa-Sicard; Harald Schmidt; Rudi Busse; Katrin Schröder; Ralf P. Brandes

A large body of literature suggest that vascular reduced nicotinamide-adenine dinucleotide phosphate (NADPH) oxidases are important sources of reactive oxygen species. Many studies, however, relied on data obtained with the inhibitor apocynin (4′-hydroxy-3′methoxyacetophenone). Because the mode of action of apocynin, however, is elusive, we determined its mechanism of inhibition on vascular NADPH oxidases. In HEK293 cells overexpressing NADPH oxidase isoforms (Nox1, Nox2, or Nox4), apocynin failed to inhibit superoxide anion generation detected by lucigenin chemiluminescence. In contrast, apocynin interfered with the detection of reactive oxygen species in assay systems selective for hydrogen peroxide or hydroxyl radicals. Importantly, apocynin interfered directly with the detection of peroxides but not superoxide, if generated by xanthine/xanthine oxidase or nonenzymatic systems. In leukocytes, apocynin is a prodrug that is activated by myeloperoxidase, a process that results in the formation of apocynin dimers. Endothelial cells and smooth muscle cells failed to form these dimers and, therefore, are not able to activate apocynin. Dimer formation was, however, observed in Nox-overexpressing HEK293 cells when myeloperoxidase was supplemented. As a consequence, apocynin should only inhibit NADPH oxidase in leukocytes, whereas in vascular cells, the compound could act as an antioxidant. Indeed, in vascular smooth muscle cells, the activation of the redox-sensitive kinases p38-mitogen-activate protein kinase, Akt, and extracellular signal–regulated kinase 1/2 by hydrogen peroxide and by the intracellular radical generator menadione was prevented in the presence of apocynin. These observations indicate that apocynin predominantly acts as an antioxidant in endothelial cells and vascular smooth muscle cells and should not be used as an NADPH oxidase inhibitor in vascular systems.


Circulation Research | 2012

Nox4 Is a Protective Reactive Oxygen Species Generating Vascular NADPH Oxidase

Katrin Schröder; Min Zhang; Sebastian Benkhoff; Anja Mieth; Rainer U. Pliquett; Judith Kosowski; Christoph Kruse; Peter Luedike; Norbert Weissmann; Stefanie Dimmeler; Ajay M. Shah; Ralf P. Brandes

Rationale: The function of Nox4, a source of vascular H2O2, is unknown. Other Nox proteins were identified as mediators of endothelial dysfunction. Objective: We determined the function of Nox4 in situations of increased stress induced by ischemia or angiotensin II with global and tamoxifen-inducible Nox4−/− mice. Methods and Results: Nox4 was highly expressed in the endothelium and contributed to H2O2 formation. Nox4−/− mice exhibited attenuated angiogenesis (femoral artery ligation) and PEG-catalase treatment in control mice had a similar effect. Tube formation in cultured Nox4−/− lung endothelial cells (LECs) was attenuated and restored by low concentrations of H2O2, whereas PEG-catalase attenuated tube formation in control LECs. Angiotensin II infusion was used as a model of oxidative stress. Compared to wild-type, aortas from inducible Nox4-deficient animals had development of increased inflammation, media hypertrophy, and endothelial dysfunction. Mechanistically, loss of Nox4 resulted in reduction of endothelial nitric oxide synthase expression, nitric oxide production, and heme oxygenase-1 (HO-1) expression, which was associated with apoptosis and inflammatory activation. HO-1 expression is controlled by Nrf-2. Accordingly, Nox4-deficient LECs exhibited reduced Nrf-2 protein level and deletion of Nox4 reduced Nrf-2 reporter gene activity. In vivo treatment with hemin, an inducer of HO-1, blocked the vascular hypertrophy induced by Nox4 deletion in the angiotensin II infusion model and carbon monoxide, the product of HO-1, blocked the Nox4-deletion-induced apoptosis in LECs. Conclusion: Endogenous Nox4 protects the vasculature during ischemic or inflammatory stress. Different from Nox1 and Nox2, this particular NADPH oxidase therefore may have a protective vascular function.


Journal of Biological Chemistry | 2011

The E-loop Is Involved in Hydrogen Peroxide Formation by the NADPH Oxidase Nox4

Ina Takac; Katrin Schröder; Leilei Zhang; Bernard Lardy; Narayana Anilkumar; J. David Lambeth; Ajay M. Shah; Françoise Morel; Ralf P. Brandes

In contrast to the NADPH oxidases Nox1 and Nox2, which generate superoxide (O2̇̄), Nox4 produces hydrogen peroxide (H2O2). We constructed chimeric proteins and mutants to address the protein region that specifies which reactive oxygen species is produced. Reactive oxygen species were measured with luminol/horseradish peroxidase and Amplex Red for H2O2 versus L-012 and cytochrome c for O2̇̄. The third extracytosolic loop (E-loop) of Nox4 is 28 amino acids longer than that of Nox1 or Nox2. Deletion of E-loop amino acids only present in Nox4 or exchange of the two cysteines in these stretches switched Nox4 from H2O2 to O2̇̄ generation while preserving expression and intracellular localization. In the presence of an NO donor, the O2̇̄-producing Nox4 mutants, but not wild-type Nox4, generated peroxynitrite, excluding artifacts of the detection system as the apparent origin of O2̇̄. In Cos7 cells, in which Nox4 partially localizes to the plasma membrane, an antibody directed against the E-loop decreased H2O2 but increased O2̇̄ formation by Nox4 without affecting Nox1-dependent O2̇̄ formation. The E-loop of Nox4 but not Nox1 and Nox2 contains a highly conserved histidine that could serve as a source for protons to accelerate spontaneous dismutation of superoxide to form H2O2. Mutation of this but not of four other conserved histidines also switched Nox4 from H2O2 to O2̇̄ formation. Thus, H2O2 formation is an intrinsic property of Nox4 that involves its E-loop. The structure of the E-loop may hinder O2̇̄ egress and/or provide a source for protons, allowing dismutation to form H2O2.


Proceedings of the National Academy of Sciences of the United States of America | 2010

NADPH oxidase-4 mediates protection against chronic load-induced stress in mouse hearts by enhancing angiogenesis

Min Zhang; Alison C. Brewer; Katrin Schröder; Celio X.C. Santos; David Grieve; Minshu Wang; Narayana Anilkumar; Bin Yu; Xuebin Dong; Simon Walker; Ralf P. Brandes; Ajay M. Shah

Cardiac failure occurs when the heart fails to adapt to chronic stresses. Reactive oxygen species (ROS)-dependent signaling is implicated in cardiac stress responses, but the role of different ROS sources remains unclear. Here we report that NADPH oxidase-4 (Nox4) facilitates cardiac adaptation to chronic stress. Unlike other Nox proteins, Nox4 activity is regulated mainly by its expression level, which increases in cardiomyocytes under stresses such as pressure overload or hypoxia. To investigate the functional role of Nox4 during the cardiac response to stress, we generated mice with a genetic deletion of Nox4 or a cardiomyocyte-targeted overexpression of Nox4. Basal cardiac function was normal in both models, but Nox4-null animals developed exaggerated contractile dysfunction, hypertrophy, and cardiac dilatation during exposure to chronic overload whereas Nox4-transgenic mice were protected. Investigation of mechanisms underlying this protective effect revealed a significant Nox4-dependent preservation of myocardial capillary density after pressure overload. Nox4 enhanced stress-induced activation of cardiomyocyte hypoxia inducible factor 1 and the release of vascular endothelial growth factor, resulting in increased paracrine angiogenic activity. These data indicate that cardiomyocyte Nox4 is a unique inducible regulator of myocardial angiogenesis, a key determinant of cardiac adaptation to overload stress. Our results also have wider relevance to the use of nonspecific antioxidant approaches in cardiac disease and may provide an explanation for the failure of such strategies in many settings.


Free Radical Biology and Medicine | 2010

NADPH oxidases in cardiovascular disease

Ralf P. Brandes; Norbert Weissmann; Katrin Schröder

Reactive oxygen species (ROS) contribute to several aspects of vascular diseases including ischemia-reperfusion injury, scavenging of nitric oxide, or stimulation of inflammation and hypertrophy. NADPH oxidases of the Nox family are differentially expressed in the cardiovascular system, induced or activated by cardiovascular risk factors and importantly contribute to the oxidative burden of vascular diseases. Moreover, NADPH oxidase-derived ROS are important signaling molecules under physiological conditions. In this article, the current knowledge on NADPH oxidase expression, activation, and signaling in the cardiovascular system as well as the impact of risk factors on the function of these proteins will be reviewed. Finally, the contribution of NADPH oxidases to the predominant cardiovascular diseases will be discussed.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2009

Nox4 Acts as a Switch Between Differentiation and Proliferation in Preadipocytes

Katrin Schröder; Katalin Wandzioch; Ina Helmcke; Ralf P. Brandes

Objective—Insulin promotes differentiation of preadipocytes into adipocytes. Insulin also stimulates reactive oxygen species (ROS) production, and the NADPH oxidases Nox1 and Nox4 are important sources of ROS. We determined in human and mouse preadipocytes whether Nox proteins contribute to ROS formation and differentiation in response to insulin. Methods and Results—The expression of Nox1 and Nox4 was increased during insulin-induced differentiation, and insulin increased ROS production. SiRNA against Nox4 but not Nox1 inhibited insulin-induced differentiation and ROS production but promoted proliferation. Nox4 overexpression yielded the opposite effect. As observed by siRNA and overexpression, Nox4 controlled the expression of MAP kinase phosphatase-1 (MKP-1), which reduces insulin-induced ERK1/2 activation. Consequently, downregulation of Nox4 promoted ERK1/2 signaling: Proliferation was increased and through phosphorylation of the inhibitory site serine612, ERK1/2 inhibited the activation of the insulin-receptor substrate-1 (IRS-1) and thereby prevented differentiation in response to insulin. Inhibition of ERK1/2 or overexpression of MPK-1 promoted insulin-induced differentiation. Accordingly, insulin-induced proliferation was enhanced by siRNA against MKP-1, whereas inhibition of ERK1/2 or overexpression of MKP-1 attenuated proliferation. Conclusions—Nox4 acts as a switch from insulin-induced proliferation to differentiation by controlling MKP-1 expression, which limits ERK1/2 signaling.


Free Radical Biology and Medicine | 2012

Liver fibrosis and hepatocyte apoptosis are attenuated by GKT137831 a novel NOX4/NOX1 inhibitor in vivo

Joy X. Jiang; Xiangling Chen; Nobuko Serizawa; Cedric Szyndralewiez; Patrick Page; Katrin Schröder; Ralf P. Brandes; Sridevi Devaraj; Natalie J. Török

Reactive oxygen species (ROS) play a key role in chronic liver injury and fibrosis. Homologs of NADPH oxidases (NOXs) are major sources of ROS, but the exact role of the individual homologs in liver disease is unknown. Our goal was to determine the role of NOX4 in liver fibrosis induced by bile duct ligation (BDL) with the aid of the pharmacological inhibitor GKT137831, and genetic deletion of NOX4 in mice. GKT137831 was either applied for the full term of BDL (preventive arm) or started at 10 day postoperatively (therapeutic arm). Primary hepatic stellate cells (HSC) from control mice with and without BDL were analyzed and the effect of NOX4 inhibition on HSC activation was also studied. FasL or TNFα/actinomycin D-induced apoptosis was studied in wild-type and NOX4(-/-) hepatocytes. NOX4 was upregulated by a TGF-β/Smad3-dependent mechanism in HSC. Downregulation of NOX4 decreased ROS production and the activation of NOX4(-/-) HSC was attenuated. NOX4(-/-) hepatocytes were more resistant to FasL or TNFα/actinomycin D-induced apoptosis. Similarly, after pharmacological NOX4 inhibition, ROS production, the expression of fibrogenic markers, and hepatocyte apoptosis were reduced. NOX4 was expressed in human livers with stage 2-3 autoimmune hepatitis. Fibrosis was attenuated by the genetic deletion of NOX4. BDL mice gavaged with GKT137831 in the preventive or the therapeutic arm displayed less ROS production, significantly attenuated fibrosis, and decreased hepatocyte apoptosis. In conclusion, NOX4 plays a key role in liver fibrosis. GKT137831 is a potent inhibitor of fibrosis and hepatocyte apoptosis; therefore, it is a promising therapeutic agent for future translational studies.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2007

Nox1 Mediates Basic Fibroblast Growth Factor-Induced Migration of Vascular Smooth Muscle Cells

Katrin Schröder; Ina Helmcke; Katalin Palfi; Karl-Heinz Krause; Rudi Busse; Ralf P. Brandes

Objectives—Basic fibroblast growth factor (bFGF) stimulates vascular smooth muscle cell (SMC) migration. We determined whether bFGF increases SMC reactive oxygen-species (ROS) and studied the role of ROS for SMC migration. Methods and Results—bFGF rapidly increased rat SMC ROS formation and migration through pathways sensitive to inhibition of NADPH oxidases, PI3-kinase, protein kinase C, and Rac-1. SiRNA directed against the NADPH oxidase Nox4 impaired basal but not bFGF-induced ROS formation and did not affect migration. In contrast, siRNA against Nox1 blocked the agonist-induced ROS generation as well as the bFGF-induced migration. Agonist-induced migration was also attenuated in SMC derived from Nox1 y/− mice and transduction of Nox1 restored normal migration. Likewise, SMC outgrowth in response to bFGF was attenuated in aortic segments from Nox1 y/− mice as compared with Nox1 y/+ mice. bFGF activated JNK but not Src in a Nox1-dependent manner. Consequently, phosphorylation of the adaptor protein paxillin, which is central for migration and secretion of matrix-metalloproteinases, were dependent on Nox1 as well as JNK but not Src. Conclusions—These data demonstrate that bFGF activates the Nox1-containing NADPH oxidase and that bFGF through a pathway involving ROS and JNK stimulates SMC migration.


Circulation Research | 2009

NADPH Oxidase Nox2 Is Required for Hypoxia-Induced Mobilization of Endothelial Progenitor Cells

Katrin Schröder; Andreas Kohnen; Alexandra Aicher; Elisa A. Liehn; Tom Büchse; Stefan Stein; Christian Weber; Stefanie Dimmeler; Ralf P. Brandes

Rationale: Endothelial progenitor cells (EPCs, defined as sca-1+flk-1+lin- mononuclear blood cells) contribute to vascular repair. The role of hypoxia and reactive oxygen species (ROS) in mobilization and function of these cells is incompletely understood. Objective: We studied the contribution of the NADPH oxidase Nox2, an important vascular source of ROS in this context. Methods and Results: Hypoxia (10% oxygen) induced the mobilization of EPCs in wild-type (WT) and Nox1 but not in Nox2 knockout (Nox2y/−) mice. As erythropoietin (EPO) is known to induce EPC mobilization, we focused on this hormone. EPO induced the mobilization of EPCs in WT and Nox1y/− but not Nox2y/− animals. Transplantation of bone marrow from Nox2y/− mice into WT-mice blocked mobilization in response to hypoxia and EPO, whereas transplantation of WT bone marrow into Nox2y/− mice restored mobilization. Reendothelialization of the injured mouse carotid artery was enhanced by hypoxia as well as by EPO, and this effect was not observed in Nox2y/− mice or after transplantation of Nox2y/− bone marrow. In cultured EPCs from WT but not Nox2y/− mice, EPO induced ROS production, migration, and proliferation. EPO signaling involves the STAT5 transcription factor. EPO-induced STAT5-dependent reporter gene expression was absent in Nox2-deficient cells. siRNA against the redox-sensitive phosphatase SHP-2 restored EPO-mediated STAT5 induction and inhibition of SHP-2 restored EPO-induced migration in Nox2-deficient cells Conclusions: We conclude that Nox2-derived ROS inactivate SHP-2 and thereby facilitate EPO signaling in EPCs to promote hypoxia-induced mobilization and vascular repair by these cells.


Current Hypertension Reports | 2012

The Nox Family of NADPH Oxidases: Friend or Foe of the Vascular System?

Ina Takac; Katrin Schröder; Ralf P. Brandes

NADPH (nicotinamide adenine dinucleotide phosphate) oxidases are important sources of reactive oxygen species (ROS). In the vascular system, ROS can have both beneficial and detrimental effects. Under physiologic conditions, ROS are involved in signaling pathways that regulate vascular tone as well as cellular processes like proliferation, migration and differentiation. However, high doses of ROS, which are produced after induction or activation of NADPH oxidases in response to cardiovascular risk factors and inflammation, contribute to the development of endothelial dysfunction and vascular disease. In vascular cells, the NADPH oxidase isoforms Nox1, Nox2, Nox4, and Nox5 are expressed, which differ in their activity, response to stimuli, and the type of ROS released. This review focuses on the specific role of different NADPH oxidase isoforms in vascular physiology and their potential contributions to vascular diseases.

Collaboration


Dive into the Katrin Schröder's collaboration.

Top Co-Authors

Avatar

Ralf P. Brandes

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Flavia Rezende

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Min Zhang

King's College London

View shared research outputs
Top Co-Authors

Avatar

Oliver Löwe

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Sebastian Benkhoff

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Rudi Busse

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Valeska Helfinger

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Franziska Moll

Goethe University Frankfurt

View shared research outputs
Researchain Logo
Decentralizing Knowledge