Network


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

Hotspot


Dive into the research topics where Andreas Üllen is active.

Publication


Featured researches published by Andreas Üllen.


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.


Neuroscience Letters | 2013

25-Hydroxycholesterol regulates cholesterol homeostasis in the murine CATH.a neuronal cell line

Sabine Waltl; Jay V. Patankar; Günter Fauler; Christoph Nusshold; Andreas Üllen; Gerald Eibinger; Andrea Wintersperger; Dagmar Kratky; Ernst Malle; Wolfgang Sattler

Highlights ► Peripheral LPS induces cholesterol 25-hydroxylase expression in mouse brain. ► 25-Hydroxycholesterol upregulates LXR target genes in a mouse neuronal cell line. ► 25-Hydroxycholesterol downregulates neuronal SREBP2-dependent transcription. ► 25-Hydroxycholesterol attenuates neuronal cholesterol biosynthesis.


Free Radical Biology and Medicine | 2016

Assessment of electrophile damage in a human brain endothelial cell line utilizing a clickable alkyne analog of 2-chlorohexadecanal

Christoph Nusshold; Andreas Üllen; Nora Kogelnik; Eva Bernhart; Helga Reicher; Ioanna Plastira; Toma N. Glasnov; Klaus Zangger; Gerald N. Rechberger; Manfred Kollroser; Günter Fauler; Heimo Wolinski; Babette B. Weksler; Ignacio A. Romero; Sepp D. Kohlwein; Pierre-Olivier Couraud; Ernst Malle; Wolfgang Sattler

Peripheral leukocytes aggravate brain damage by releasing cytotoxic mediators that compromise blood-brain barrier function. One of the oxidants released by activated leukocytes is hypochlorous acid (HOCl) that is formed via the myeloperoxidase-H2O2-chloride system. The reaction of HOCl with the endogenous plasmalogen pool of brain endothelial cells results in the generation of 2-chlorohexadecanal (2-ClHDA), a toxic, lipid-derived electrophile that induces blood-brain barrier dysfunction in vivo. Here, we synthesized an alkynyl-analog of 2-ClHDA, 2-chlorohexadec-15-yn-1-al (2-ClHDyA) to identify potential protein targets in the human brain endothelial cell line hCMEC/D3. Similar to 2-ClHDA, 2-ClHDyA administration reduced cell viability/metabolic activity, induced processing of pro-caspase-3 and PARP, and led to endothelial barrier dysfunction at low micromolar concentrations. Protein-2-ClHDyA adducts were fluorescently labeled with tetramethylrhodamine azide (N3-TAMRA) by 1,3-dipolar cycloaddition in situ, which unveiled a preferential accumulation of 2-ClHDyA adducts in mitochondria, the Golgi, endoplasmic reticulum, and endosomes. Thirty-three proteins that are subject to 2-ClHDyA-modification in hCMEC/D3 cells were identified by mass spectrometry. Identified proteins include cytoskeletal components that are central to tight junction patterning, metabolic enzymes, induction of the oxidative stress response, and electrophile damage to the caveolar/endosomal Rab machinery. A subset of the targets was validated by a combination of N3-TAMRA click chemistry and specific antibodies by fluorescence microscopy. This novel alkyne analog is a valuable chemical tool to identify cellular organelles and protein targets of 2-ClHDA-mediated damage in settings where myeloperoxidase-derived oxidants may play a disease-propagating role.


Biochemical Pharmacology | 2015

Covalent adduct formation between the plasmalogen-derived modification product 2-chlorohexadecanal and phloretin

Andreas Üllen; Christoph Nusshold; Toma N. Glasnov; Robert Saf; David Cantillo; Gerald Eibinger; Helga Reicher; Günter Fauler; Eva Bernhart; Seth Hallström; Nora Kogelnik; Klaus Zangger; C. Oliver Kappe; Ernst Malle; Wolfgang Sattler

Hypochlorous acid added as reagent or generated by the myeloperoxidase (MPO)-H2O2-Cl(-) system oxidatively modifies brain ether-phospholipids (plasmalogens). This reaction generates a sn2-acyl-lysophospholipid and chlorinated fatty aldehydes. 2-Chlorohexadecanal (2-ClHDA), a prototypic member of chlorinated long-chain fatty aldehydes, has potent neurotoxic potential by inflicting blood-brain barrier (BBB) damage. During earlier studies we could show that the dihydrochalcone-type polyphenol phloretin attenuated 2-ClHDA-induced BBB dysfunction. To clarify the underlying mechanism(s) we now investigated the possibility of covalent adduct formation between 2-ClHDA and phloretin. Coincubation of 2-ClHDA and phloretin in phosphatidylcholine liposomes revealed a half-life of 2-ClHDA of approx. 120min, decaying at a rate of 5.9×10(-3)min(-1). NMR studies and enthalpy calculations suggested that 2-ClHDA-phloretin adduct formation occurs via electrophilic aromatic substitution followed by hemiacetal formation on the A-ring of phloretin. Adduct characterization by high-resolution mass spectroscopy confirmed these results. In contrast to 2-ClHDA, the covalent 2-ClHDA-phloretin adduct was without adverse effects on MTT reduction (an indicator for metabolic activity), cellular adenine nucleotide content, and barrier function of brain microvascular endothelial cells (BMVEC). Of note, 2-ClHDA-phloretin adduct formation was also observed in BMVEC cultures. Intraperitoneal application and subsequent GC-MS analysis of brain lipid extracts revealed that phloretin is able to penetrate the BBB of C57BL/6J mice. Data of the present study indicate that phloretin scavenges 2-ClHDA, thereby attenuating 2-ClHDA-mediated brain endothelial cell dysfunction. We here identify a detoxification pathway for a prototypic chlorinated fatty aldehyde (generated via the MPO axis) that compromises BBB function in vitro and in vivo.


BMC Pharmacology | 2010

The pulmonary microvascular endothelial barrier function is controlled by the PGE2-EP4 signaling axis

Viktoria Konya; Andreas Üllen; Eva M. Sturm; Petra Luschnig; Wolfgang Sattler; Akos Heinemann

Background Endothelial cells, like gate-keepers of the vascular bed, can actively protect against the inflammatory process. Prostaglandin E2 (PGE2) could be one of the mediators that can promote the barrier function of endothelial cells. PGE2 exerts its cellular effects by binding to four different E-prostanoid receptors (EP1–4) that belong to the family of G protein-coupled receptors. This project aimed at characterizing the barrier-protective properties of PGE2 and especially EP4 receptor on human pulmonary microvascular endothelial cells (HMVEC-L).


Free Radical Biology and Medicine | 2010

Mouse brain plasmalogens are targets for hypochlorous acid-mediated modification in vitro and in vivo.

Andreas Üllen; Günter Fauler; Harald Köfeler; Sabine Waltl; Christoph Nusshold; Eva Bernhart; Helga Reicher; Hans-Jörg Leis; Andrea Wintersperger; Ernst Malle; Wolfgang Sattler


Free Radical Biology and Medicine | 2011

Corrigendum to “Mouse brain plasmalogens are targets for hypochlorous acid-mediated modification in vitro and in vivo” [Free Radic. Biol. Med. 49 (2010) 1655-1665]

Andreas Üllen; Günter Fauler; Harald Köfeler; Sabine Waltl; Christoph Nusshold; Eva Bernhart; Helga Reicher; Hans-Jörg Leis; Andrea Wintersperger; Ernst Malle; Wolfgang Sattler


/data/revues/00916749/v131i2/S0091674912007828/ | 2013

Endothelial E-type prostanoid 4 receptors promote barrier function and inhibit neutrophil trafficking

Viktoria Konya; Andreas Üllen; Nora Kampitsch; Anna Theiler; Sonia Philipose; Gerald P. Parzmair; Gunther Marsche; Bernhard A. Peskar; Rufina Schuligoi; Wolfgang Sattler; Akos Heinemann

Collaboration


Dive into the Andreas Üllen's collaboration.

Top Co-Authors

Avatar

Wolfgang Sattler

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ernst Malle

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Günter Fauler

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Helga Reicher

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

Sabine Waltl

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar

Akos Heinemann

Medical University of Graz

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hans-Jörg Leis

Medical University of Graz

View shared research outputs
Researchain Logo
Decentralizing Knowledge