Network


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

Hotspot


Dive into the research topics where Helga Reicher is active.

Publication


Featured researches published by Helga Reicher.


Gastroenterology | 2012

Alterations in Lipid Metabolism Mediate Inflammation, Fibrosis, and Proliferation in a Mouse Model of Chronic Cholestatic Liver Injury

Tarek Moustafa; Peter Fickert; Christoph Magnes; Christian Guelly; Andrea Thueringer; Saša Frank; Dagmar Kratky; Wolfgang Sattler; Helga Reicher; Frank Sinner; Judith Gumhold; Dagmar Silbert; Günter Fauler; Gerald Höfler; Achim Lass; Rudolf Zechner; Michael Trauner

BACKGROUND & AIMS The liver controls central processes of lipid and bile acid homeostasis. We aimed to investigate whether alterations in lipid metabolism contribute to the pathogenesis of chronic cholestatic liver disease in mice. METHODS We used microarray and metabolic profiling analyses to identify alterations in systemic and hepatic lipid metabolism in mice with disruption of the gene ATP-binding cassette sub-family B member 4 (Abcb4(-/-) mice), a model of inflammation-induced cholestatic liver injury, fibrosis, and cancer. RESULTS Alterations in Abcb4(-/-) mice, compared with wild-type mice, included deregulation of genes that control lipid synthesis, storage, and oxidation; decreased serum levels of cholesterol and phospholipids; and reduced hepatic long-chain fatty acyl-CoAs (LCA-CoA). Feeding Abcb4(-/-) mice the side chain-modified bile acid 24-norursodeoxycholic acid (norUDCA) reversed their liver injury and fibrosis, increased serum levels of lipids, lowered phospholipase and triglyceride hydrolase activities, and restored hepatic LCA-CoA and triglyceride levels. Additional genetic and nutritional studies indicated that lipid metabolism contributed to chronic cholestatic liver injury; crossing peroxisome proliferator-activated receptor (PPAR)-α-deficient mice with Abcb4(-/-) mice (to create double knockouts) or placing Abcb4(-/-) mice on a high-fat diet protected against liver injury, with features similar to those involved in the response to norUDCA. Placing pregnant Abcb4(-/-) mice on high-fat diets prevented liver injury in their offspring. However, fenofibrate, an activator of PPARα, aggravated liver injury in Abcb4(-/-) mice. CONCLUSIONS Alterations in lipid metabolism contribute to the pathogenesis and progression of cholestatic liver disease in mice.


Biochimica et Biophysica Acta | 2000

Lipoprotein-associated α-tocopheryl-succinate inhibits cell growth and induces apoptosis in human MCF-7 and HBL-100 breast cancer cells

Pirkko J. Pussinen; Helmut Lindner; Otto Glatter; Helga Reicher; Gerhard M. Kostner; Andrea Wintersperger; Ernst Malle; Wolfgang Sattler

K-Tocopheryl succinate (K-TS) is a potent inhibitor of tumor cell proliferation. The goal of the present study was to investigate whether and to what extent K-TS associates with plasma lipoproteins and if K-TS-enriched lipoproteins inhibit breast cancer cell growth in a manner comparable to the free drug. In vitro enrichment of human plasma revealed that K-TS readily associated with the main lipoprotein classes, findings confirmed in vivo in mice. At the highest K-TS concentrations, lipoproteins carrying 50 000 (VLDL), 5000 (LDL) and 700 (HDL) K-TS molecules per lipoprotein particle were generated. KTS enrichment generated lipoprotein particles with slightly decreased density and increased particle radius. To study whether the level of LDL-receptor (LDL-R) expression affects K-TS uptake from apoB/E containing lipoprotein particles human breast cancer cells with low (MCF-7) and normal (HBL-100) LDL-R expression were used. The uptake of free, VLDL- and (apoE-free) HDL3-associated K-TS was nearly identical for both cell lines. In contrast, uptake of LDL-associated K-TS by HBL-100 cells (normal LDL-R expression) was about twice as high as compared to MCF-7 cells (low LDL-R expression). VLDL and LDL-associated K-TS inhibited proliferation most effectively at the highest concentration of K-TS used (100% inhibition of MCF-7 growth with 20 Wg/ml of lipoprotein-associated K-TS). However, also K-TS-free VLDL and LDL inhibited HBL-100 cell proliferation up to 55%. In both cell lines, K-TS-enriched HDL3 inhibited cell growth by 40^60%. Incubation of both cell lines in the presence of free or lipoprotein-associated K-TS resulted in DNA fragmentation indicative of apoptosis. Collectively, the present findings demonstrate that: (1) K-TS readily associates with lipoproteins in vitro and in vivo; (2) the lipoprotein-enrichment efficacy was dependent on the particle size and/or the triglyceride content of the lipoprotein; (3) uptake of LDL-associated K-TS was apparently dependent on the level of LDL-R expression ; and (4) lipoproteins were efficient K-TS carriers inducing reduced cell proliferation rates and apoptosis in human breast cancer cells as observed for the free drug. fl 2000 Elsevier Science B.V. All rights reserved.


Journal of Biological Chemistry | 2011

Sequential Synthesis and Methylation of Phosphatidylethanolamine Promote Lipid Droplet Biosynthesis and Stability in Tissue Culture and in Vivo

Gerd Hörl; Andrea Wagner; Laura K. Cole; Roland Malli; Helga Reicher; Petra Kotzbeck; Harald Köfeler; Gerald Höfler; Saša Frank; Juliane G. Bogner-Strauss; Wolfgang Sattler; Dennis E. Vance; Ernst Steyrer

Triacylglycerols are stored in eukaryotic cells within lipid droplets (LD). The LD core is enwrapped by a phospholipid monolayer with phosphatidylcholine (PC), the major phospholipid, and phosphatidylethanolamine (PE), a minor component. We demonstrate that the onset of LD formation is characterized by a change in cellular PC, PE, and phosphatidylserine (PS). With induction of differentiation of 3T3-L1 fibroblasts into adipocytes, the cellular PC/PE ratio decreased concomitant with LD formation, with the most pronounced decline between confluency and day 5. The mRNA for PS synthase-1 (forms PS from PC) and PS decarboxylase (forms PE from PS) increased after day 5. Activity and protein of PE N-methyltransferase (PEMT), which produces PC by methylation of PE, are absent in 3T3-L1 fibroblasts but were induced at day 5. High fat challenge induced PEMT expression in mouse adipose tissue. PE, produced via PS decarboxylase, was the preferred substrate for methylation to PC. A PEMT-GFP fusion protein decorated the periphery of LD. PEMT knockdown in 3T3-L1 adipocytes correlated with increased basal triacylglycerol hydrolysis. Pemt−/− mice developed desensitization against adenosine-mediated inhibition of basal hydrolysis in adipose tissue, and adipocyte hypotrophy was observed in Pemt−/− animals on a high fat diet. Knock-out of PEMT in adipose tissue down-regulated PS synthase-1 mRNA, suggesting coordination between PE supply and converting pathways during LD biosynthesis. We conclude that two consecutive processes not previously related to LD biogenesis, (i) PE production via PS and (ii) PE conversion via PEMT, are implicated in LD formation and stability.


Proteomics | 2010

Lysophosphatidic acid receptor activation affects the C13NJ microglia cell line proteome leading to alterations in glycolysis, motility, and cytoskeletal architecture

Eva Bernhart; Manfred Kollroser; Gerald N. Rechberger; Helga Reicher; Akos Heinemann; Petra Schratl; Seth Hallström; Andrea Wintersperger; Christoph Nusshold; Trevor DeVaney; Klaus Zorn-Pauly; Roland Malli; Wolfgang F. Graier; Ernst Malle; Wolfgang Sattler

Microglia, the immunocompetent cells of the CNS, are rapidly activated in response to injury and microglia migration towards and homing at damaged tissue plays a key role in CNS regeneration. Lysophosphatidic acid (LPA) is involved in signaling events evoking microglia responses through cognate G protein‐coupled receptors. Here we show that human immortalized C13NJ microglia express LPA receptor subtypes LPA1, LPA2, and LPA3 on mRNA and protein level. LPA activation of C13NJ cells induced Rho and extracellular signal‐regulated kinase activation and enhanced cellular ATP production. In addition, LPA induced process retraction, cell spreading, led to pronounced changes of the actin cytoskeleton and reduced cell motility, which could be reversed by inhibition of Rho activity. To get an indication about LPA‐induced global alterations in protein expression patterns a 2‐D DIGE/LC‐ESI‐MS proteomic approach was applied. On the proteome level the most prominent changes in response to LPA were observed for glycolytic enzymes and proteins regulating cell motility and/or cytoskeletal dynamics. The present findings suggest that naturally occurring LPA is a potent regulator of microglia biology. This might be of particular relevance in the pathophysiological context of neurodegenerative disorders where LPA concentrations can be significantly elevated in the CNS.


Free Radical Biology and Medicine | 2010

Hypochlorite modification of sphingomyelin generates chlorinated lipid species that induce apoptosis and proteome alterations in dopaminergic PC12 neurons in vitro.

Christoph Nusshold; Manfred Kollroser; Harald Köfeler; Gerald N. Rechberger; Helga Reicher; Andreas Üllen; Eva Bernhart; Sabine Waltl; Ingrid Kratzer; Albin Hermetter; Hubert Hackl; Zlatko Trajanoski; Andelko Hrzenjak; Ernst Malle; Wolfgang Sattler

Recent observations link myeloperoxidase (MPO) activation to neurodegeneration. In multiple sclerosis MPO is present in areas of active demyelination where the potent oxidant hypochlorous acid (HOCl), formed by MPO from H(2)O(2) and chloride ions, could oxidatively damage myelin-associated lipids. The purpose of this study was (i) to characterize reaction products of sphingomyelin (SM) formed in response to modification by HOCl, (ii) to define the impact of exogenously added SM and HOCl-modified SM (HOCl-SM) on viability parameters of a neuronal cell line (PC12), and (iii) to study alterations in the PC12 cell proteome in response to SM and HOCl-SM. MALDI-TOF-MS analyses revealed that HOCl, added as reagent or generated enzymatically, transforms SM into chlorinated species. On the cellular level HOCl-SM but not SM induced the formation of reactive oxygen species. HOCl-SM induced severely impaired cell viability, dissipation of the mitochondrial membrane potential, and activation of caspase-3 and DNA damage. Proteome analyses identified differential expression of specific subsets of proteins in response to SM and HOCl-SM. Our results demonstrate that HOCl modification of SM results in the generation of chlorinated lipid species with potent neurotoxic properties. Given the emerging connections between the MPO-H(2)O(2)-chloride axis and neurodegeneration, this chlorinating pathway might be implicated in neuropathogenesis.


Free Radical Biology and Medicine | 2012

Phloretin ameliorates 2-chlorohexadecanal-mediated brain microvascular endothelial cell dysfunction in vitro

Andreas Üllen; Günter Fauler; Eva Bernhart; Christoph Nusshold; Helga Reicher; Hans-Jörg Leis; Ernst Malle; Wolfgang Sattler

2-Chlorohexadecanal (2-ClHDA), a chlorinated fatty aldehyde, is formed via attack on ether-phospholipids by hypochlorous acid (HOCl) that is generated by the myeloperoxidase–hydrogen peroxide–chloride system of activated leukocytes. 2-ClHDA levels are elevated in atherosclerotic lesions, myocardial infarction, and neuroinflammation. Neuroinflammatory conditions are accompanied by accumulation of neutrophils (an ample source of myeloperoxidase) in the brain. Microvessel damage by inflammatory mediators and/or reactive oxidants can induce blood–brain barrier (BBB) dysfunction, a pathological condition leading to cerebral edema, brain hemorrhage, and neuronal death. In this in vitro study we investigated the impact of 2-ClHDA on brain microvascular endothelial cells (BMVEC), which constitute the morphological basis of the BBB. We show that exogenously added 2-ClHDA is subject to rapid uptake and metabolism by BMVEC. Using C16 structural analogues of 2-ClHDA we found that the cytotoxic potential decreases in the following order: 2-ClHDA>hexadecanal>palmitic acid>2-ClHDA-dimethylacetal. 2-ClHDA induces loss of barrier function, mitochondrial dysfunction, apoptosis via activation of caspase 3, and altered intracellular redox balance. Finally we investigated potential protective effects of several natural polyphenols on in vitro BBB function. Of the compounds tested, phloretin almost completely abrogated 2-ClHDA-induced BMVEC barrier dysfunction and cell death. These data suggest that 2-ClHDA has the potential to induce BBB breakdown under inflammatory conditions and that phloretin confers protection in this experimental setting.


PLOS ONE | 2013

Myeloperoxidase-Derived Oxidants Induce Blood-Brain Barrier Dysfunction In Vitro and In Vivo

Andreas Üllen; Evelin Singewald; Viktoria Konya; Günter Fauler; Helga Reicher; Christoph Nusshold; Astrid Hammer; Dagmar Kratky; Akos Heinemann; Peter Holzer; Ernst Malle; Wolfgang Sattler

Peripheral leukocytes can exacerbate brain damage by release of cytotoxic mediators that disrupt blood-brain barrier (BBB) function. One of the oxidants released by activated leukocytes is hypochlorous acid (HOCl) formed via the myeloperoxidase (MPO)-H2O2-Cl− system. In the present study we examined the role of leukocyte activation, leukocyte-derived MPO and MPO-generated oxidants on BBB function in vitro and in vivo. In a mouse model of lipopolysaccharide (LPS)-induced systemic inflammation, neutrophils that had become adherent released MPO into the cerebrovasculature. In vivo, LPS-induced BBB dysfunction was significantly lower in MPO-deficient mice as compared to wild-type littermates. Both, fMLP-activated leukocytes and the MPO-H2O2-Cl− system inflicted barrier dysfunction of primary brain microvascular endothelial cells (BMVEC) that was partially rescued with the MPO inhibitor 4-aminobenzoic acid hydrazide. BMVEC treatment with the MPO-H2O2-Cl− system or activated neutrophils resulted in the formation of plasmalogen-derived chlorinated fatty aldehydes. 2-chlorohexadecanal (2-ClHDA) severely compromised BMVEC barrier function and induced morphological alterations in tight and adherens junctions. In situ perfusion of rat brain with 2-ClHDA increased BBB permeability in vivo. 2-ClHDA potently activated the MAPK cascade at physiological concentrations. An ERK1/2 and JNK antagonist (PD098059 and SP600125, respectively) protected against 2-ClHDA-induced barrier dysfunction in vitro. The current data provide evidence that interference with the MPO pathway could protect against BBB dysfunction under (neuro)inflammatory conditions.


Journal of Neuroinflammation | 2016

1-Oleyl-lysophosphatidic acid (LPA) promotes polarization of BV-2 and primary murine microglia towards an M1-like phenotype

Ioanna Plastira; Eva Bernhart; Madeleine Goeritzer; Helga Reicher; Vishwanath Bhat Kumble; Nora Kogelnik; Andrea Wintersperger; Astrid Hammer; Stefanie Schlager; Katharina Jandl; Akos Heinemann; Dagmar Kratky; Ernst Malle; Wolfgang Sattler

BackgroundMicroglia, the immunocompetent cells of the CNS, rapidly respond to brain injury and disease by altering their morphology and phenotype to adopt an activated state. Microglia can exist broadly between two different states, namely the classical (M1) and the alternative (M2) phenotype. The first is characterized by the production of pro-inflammatory cytokines/chemokines and reactive oxygen and/or nitrogen species. In contrast, alternatively activated microglia are typified by an anti-inflammatory phenotype supporting wound healing and debris clearance. The objective of the present study was to determine the outcome of lysophosphatidic acid (LPA)-mediated signaling events on microglia polarization.MethodsLPA receptor expression and cyto-/chemokine mRNA levels in BV-2 and primary murine microglia (PMM) were determined by qPCR. M1/M2 marker expression was analyzed by Western blotting, immunofluorescence microscopy, or flow cytometry. Cyto-/chemokine secretion was quantitated by ELISA.ResultsBV-2 cells express LPA receptor 2 (LPA2), 3, 5, and 6, whereas PMM express LPA1, 2, 4, 5, and 6. We show that LPA treatment of BV-2 and PMM leads to a shift towards a pro-inflammatory M1-like phenotype. LPA treatment increased CD40 and CD86 (M1 markers) and reduced CD206 (M2 marker) expression. LPA increased inducible nitric oxide synthase (iNOS) and COX-2 levels (both M1), while the M2 marker Arginase-1 was suppressed in BV-2 cells. Immunofluorescence studies (iNOS, COX-2, Arginase-1, and RELMα) extended these findings to PMM. Upregulation of M1 markers in BV-2 and PMM was accompanied by increased cyto-/chemokine transcription and secretion (IL-1β, TNFα, IL-6, CCL5, and CXCL2). The pharmacological LPA5 antagonist TCLPA5 blunted most of these pro-inflammatory responses.ConclusionsLPA drives BV-2 and PMM towards a pro-inflammatory M1-like phenotype. Suppression by TCLPA5 indicates that the LPA/LPA5 signaling axis could represent a potential pharmacological target to interfere with microglia polarization in disease.


Biochemical Pharmacology | 2015

Interference with distinct steps of sphingolipid synthesis and signaling attenuates proliferation of U87MG glioma cells

Eva Bernhart; Sabine Damm; Andrea Wintersperger; Christoph Nusshold; Anna Martina Brunner; Ioanna Plastira; Gerald N. Rechberger; Helga Reicher; Christian Wadsack; Andreas Zimmer; Ernst Malle; Wolfgang Sattler

Graphical abstract


Biochemical Pharmacology | 2017

Dipeptidyl peptidase-4 independent cardiac dysfunction links saxagliptin to heart failure

Chintan N. Koyani; Ewald Kolesnik; Gerald Wölkart; Niroj Shrestha; Susanne Scheruebel; Christopher Trummer; Klaus Zorn-Pauly; Astrid Hammer; Petra Lang; Helga Reicher; Heinrich Maechler; Klaus Groschner; Bernd Mayer; Peter P. Rainer; Harald Sourij; Wolfgang Sattler; Ernst Malle; Brigitte Pelzmann; Dirk von Lewinski

ABSTRACT Saxagliptin treatment has been associated with increased rate of hospitalization for heart failure in type 2 diabetic patients, though the underlying mechanism(s) remain elusive. To address this, we assessed the effects of saxagliptin on human atrial trabeculae, guinea pig hearts and cardiomyocytes. We found that the primary target of saxagliptin, dipeptidyl peptidase‐4, is absent in cardiomyocytes, yet saxagliptin internalized into cardiomyocytes and impaired cardiac contractility via inhibition of the Ca2+/calmodulin‐dependent protein kinase II‐phospholamban‐sarcoplasmic reticulum Ca2+‐ATPase 2a axis and Na+‐Ca2+ exchanger function in Ca2+ extrusion. This resulted in reduced sarcoplasmic reticulum Ca2+ content, diastolic Ca2+ overload, systolic dysfunction and impaired contractile force. Furthermore, saxagliptin reduced protein kinase C‐mediated delayed rectifier K+ current that prolonged action potential duration and consequently QTc interval. Importantly, saxagliptin aggravated pre‐existing cardiac dysfunction induced by ischemia/reperfusion injury. In conclusion, our novel results provide mechanisms for the off‐target deleterious effects of saxagliptin on cardiac function and support the outcome of SAVOR‐TIMI 53 trial that linked saxagliptin with the risk of heart failure.

Collaboration


Dive into the Helga Reicher's collaboration.

Top Co-Authors

Avatar

Wolfgang Sattler

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Ernst Malle

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Eva Bernhart

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Günter Fauler

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Andreas Üllen

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Astrid Hammer

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Dagmar Kratky

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge