Lars-Ove Brandenburg
RWTH Aachen University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Lars-Ove Brandenburg.
Annals of the Rheumatic Diseases | 2011
Christoph Jan Wruck; Athanassios Fragoulis; Agata Gurzynski; Lars-Ove Brandenburg; Yuet Wai Kan; Kaimin Chan; Joachim Hassenpflug; Sandra Freitag-Wolf; Deike Varoga; Sebastian Lippross; Thomas Pufe
Objectives Increasing evidence suggests that oxidative stress may play a key role in joint destruction due to rheumatoid arthritis (RA). The aim of this study was to elucidate the role of nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor that maintains the cellular defence against oxidative stress, in RA. Methods The activation status of Nrf2 was assessed in synovial tissue from patients with RA using immunohistochemistry. Antibody-induced arthritis (AIA) was induced in Nrf2-knockout and Nrf2-wild-type control mice. The severity of cartilage destruction was evaluated using a damage score. The extent of oxidative stress, the activation state of Nrf2 and the expression level of Nrf2 target genes were analysed by immunhistological staining. The expression of vascular endothelial growth factor (VEGF)-A was examined on mRNA and protein using the Luminex technique. A Xenogen imaging system was used to measure Nrf2 activity in an antioxidant response element-luciferase transgenic mouse during AIA. Results Nrf2 was activated in the joints of arthritic mice and of patients with RA. Nrf2-knockout mice had more severe cartilage injuries and more oxidative damage, and the expression of Nrf2 target genes was enhanced in Nrf2-wild-type but not in knockout mice during AIA. Both VEGF-A mRNA and protein expression was upregulated in Nrf2-knockout mice during AIA. An unexpected finding was the number of spontaneously fractured bones in Nrf2-knockout mice with AIA. Conclusion These results provide strong evidence that oxidative stress is significantly involved in cartilage degradation in experimental arthritis, and indicate that the presence of a functional Nrf2 gene is a major requirement for limiting cartilage destruction.
Molecular Pharmacology | 2008
Christoph Jan Wruck; Mario E. Götz; Thomas Herdegen; Deike Varoga; Lars-Ove Brandenburg; Thomas Pufe
One hallmark of Alzheimers disease is the accumulation of amyloid β-peptide (AP), which can initiate a cascade of oxidative events that may result in neuronal death. Because nuclear factor erythroid 2-related factor 2 (Nrf2) is the major regulator for a battery of genes encoding detoxifying and antioxidative enzymes via binding to the antioxidant response element (ARE), it is of great interest to find nontoxic activators of Nrf2 rendering neuronal cells more resistant to AP toxicity. Using ARE-luciferase assay and Western blot, we provide evidence that the kavalactones methysticin, kavain, and yangonin activate Nrf2 time- and dose-dependently in neural PC-12 and astroglial C6 cells and thereby up-regulate cytoprotective genes. Viability and cytotoxicity assays demonstrate that Nrf2 activation is able to protect neural cells from amyloid β-(1-42) induced neurotoxicity. Down-regulation of Nrf2 by small hairpin RNA as well as extracellular signal-regulated kinase 1/2 inhibition abolishes cytoprotection. We further give evidence that kavalactone-mediated Nrf2 activation is not dependent on oxidative stress production. Our results demonstrate that kavalactones attenuate amyloid β-peptide toxicity by inducing protective gene expression mediated by Nrf2 activation in vitro. These findings indicate that the use of purified kavalactones might be considered as an adjunct therapeutic strategy to combat neural demise in Alzheimer disease and other oxidative stress-related diseases.
Inflammation Research | 2010
Lars-Ove Brandenburg; Markus Kipp; Ralph Lucius; Thomas Pufe; Christoph Jan Wruck
Objective and designThe aim of this study was to investigate the signal transduction pathways involved in sulforaphane (SF) mediated inhibition of the inflammatory response to lipopolysaccharide (LPS). Additionally, we investigated the effects of SF and LPS on the activity of Nrf2.MaterialPrimary rat microglia and the murine microglia cell line BV2 were used.TreatmentCells were treated with LPS with or without SF.MethodsCell viability was measured via WST-assay. Real-time RT-PCR was performed to analyze cytokine mRNA levels. The nitric oxide (NO) release was measured in LPS-stimulated microglia. The induction of various signal transduction pathways and Nrf2 was determined by Western blotting. NF-κB and AP-1 activation was measured by dual luciferase assay.ResultsWe showed that SF attenuates the LPS-induced increase of IL-1β, IL-6, and TNF-α expression in microglia. In addition, SF significantly decreases the NO in a concentration-dependent manner. SF inhibits LPS-stimulated ERK1/2 and JNK phosphorylation and thereby inhibits the LPS-induced activation of NF-κB- and activator protein-1 (AP-1). Moreover, SF and LPS together are able to induce Nrf2 activation.ConclusionsWe showed that SF, and also LPS by itself, are able to activate the cell’s defence against oxidative and electrophilic stress. We conclude that SF could be a candidate agent for anti-inflammatory treatment of the central nervous system.
Histochemistry and Cell Biology | 2011
Mersedeh Tohidnezhad; Deike Varoga; Christoph Jan Wruck; Lars-Ove Brandenburg; Andreas Seekamp; Mehdi Shakibaei; Tolga Taha Sönmez; Thomas Pufe; Sebastian Lippross
Little is know about the pathophysiology of acute and degenerative tendon injuries. Although most lesions are uncomplicated, treatment is long and unsatisfactory in a considerable number of cases. Besides the common growth factors that were shown to be relevant for tendon integrity more recently protection against oxidative stress was shown to promote tendon healing. To improve tendon regeneration, many have advocated the use of platelet-rich plasma (PRP), a thrombocyte concentrate that can serve as an autologous source of growth factors. In this study, we investigated the effect of platelet-released growth factors (PRGF) on tenocytes. Tenocytes were isolated from the Achilles tendon of postnatal rats. Tenocyte cell cultures were stimulated with PRGF. We used a CyQuant assay and WST assay to analyse tendon cell growth and viability in different concentrations of PRGF. Migration and proliferation of cells grown in PRGF were assessed by a scratch test. A dual-luciferase assay was used to demonstrate the activation of the anti-oxidant response element (ARE) in tenocytes. A positive effect of PRGF could be shown on tendon cell growth and migratory capacity. PRGF activated the Nrf2–ARE pathway in a dose-dependent manner. Here, we provide evidence of a biological effect of PRGF on tenocytes by the promotion of tenocyte growth and activation of the Nrf2–ARE pathway. This is a novel aspect of the action of platelet concentrates on tendon growth.
Journal of Biological Chemistry | 2011
Christoph Jan Wruck; Konrad L. Streetz; Goran Pavic; Mario E. Götz; Mersedeh Tohidnezhad; Lars-Ove Brandenburg; Deike Varoga; Oliver Eickelberg; Thomas Herdegen; Christian Trautwein; Kaimin Cha; Yuet Wai Kan; Thomas Pufe
IL-6 gene expression is controlled by a promoter region containing multiple regulatory elements such as NF-κB, NF-IL6, CRE, GRE, and TRE. In this study, we demonstrated that TRE, found within the IL-6 promoter, is embedded in a functional antioxidant response element (ARE) matching an entire ARE consensus sequence. Further, point mutations of the ARE consensus sequence in the IL-6 promoter construct selectively eliminate ARE but not TRE activity. Nrf2 is a redox-sensitive transcription factor which provides cytoprotection against electrophilic and oxidative stress and is the most potent activator of ARE-dependent transcription. Using Nrf2 knock-out mice we demonstrate that Nrf2 is a potent activator of IL-6 gene transcription in vivo. Moreover, we show evidence that Nrf2 is the transcription factor that activates IL6 expression in a cholestatic hepatitis mouse model. Our findings suggest a possible role of IL-6 in oxidative stress defense and also give indication about an important function for Nrf2 in the regulation of hematopoietic and inflammatory processes.
Journal of Neuropathology and Experimental Neurology | 2008
Lars-Ove Brandenburg; Deike Varoga; Nicoletta Nicolaeva; Stephen L. Leib; Henrik Wilms; Rainer Podschun; Christoph Jan Wruck; Jens-Michael Schröder; Thomas Pufe; Ralph Lucius
Abstract Antimicrobial peptides are intrinsic to the innate immune system in many organ systems, but little is known about their expression in the central nervous system. We examined cerebrospinal fluid (CSF) and serum from patients with active bacterial meningitis to assess antimicrobial peptides and possible bactericidal properties of the CSF. We found antimicrobial peptides (human cathelicidin LL-37) in the CSF of patients with bacterial meningitis but not in control CSF. We next characterized the expression, secretion, and bactericidal properties of rat cathelin-related antimicrobial peptide, the homologue of the human LL-37, in rat astrocytes and microglia after incubation with different bacterial components. Using real-time polymerase chain reaction and Western blotting, we determined that supernatants from both astrocytes and microglia incubated with bacterial component supernatants had antimicrobial activity. The expression of rat cathelin-related antimicrobial peptide in rat glial cells involved different signal transduction pathways and was induced by the inflammatory cytokines interleukin 1&bgr; and tumor necrosis factor. In an experimental model of meningitis, infant rats were intracisternally infected with Streptococcus pneumoniae, and rat cathelin-related antimicrobial peptide was localized in glia, choroid plexus, and ependymal cells by immunohistochemistry. Together, these results suggest that cathelicidins produced by glia and other cells play an important part in the innate immune response against pathogens in central nervous system bacterial infections.
Molecular Neurodegeneration | 2012
Alexander Slowik; Julika Merres; Anne Elfgen; Sandra Jansen; Fabian Mohr; Christoph Jan Wruck; Thomas Pufe; Lars-Ove Brandenburg
BackgroundRecent studies suggest that the chemotactic G-protein-coupled-receptor (GPCR) formyl-peptide-receptor-like-1 (FPRL1) and the receptor-for-advanced-glycation-end-products (RAGE) play an important role in the inflammatory response involved in neurodegenerative disorders such as Alzheimer’s disease (AD).Therefore, the expression and co-localisation of mouse formyl peptide receptor (mFPR) 1 and 2 as well as RAGE in an APP/PS1 transgenic mouse model using immunofluorescence and real-time RT-PCR were analysed. The involvement of rat or human FPR1/FPRL1 (corresponds to mFPR1/2) and RAGE in amyloid-β 1–42 (Aβ1-42)-induced signalling were investigated by extracellular signal regulated kinase 1/2 (ERK1/2) phosphorylation. Furthermore, the cAMP level in primary rat glial cells (microglia and astrocytes) and transfected HEK 293 cells was measured. Formyl peptide receptors and RAGE were inhibited by a small synthetic antagonist WRW4 and an inactive receptor variant delta-RAGE, lacking the intracytoplasmatic domains.ResultsWe demonstrated a strong increase of mFPR1/2 and RAGE expression in the cortex and hippocampus of APP/PS1 transgenic mice co-localised to the glial cells. In addition, the Aβ1-42-induced signal transduction is dependant on FPRL1, but also on FPR1. For the first time, we have shown a functional interaction between FPRL1/FPR1 and RAGE in RAGE ligands S100B- or AGE-mediated signalling by ERK1/2 phosphorylation and cAMP level measurement. In addition a possible physical interaction between FPRL1 as well as FPR1 and RAGE was shown with co-immunoprecipitation and fluorescence microscopy.ConclusionsThe results suggest that both formyl peptide receptors play an essential role in Aβ1-42-induced signal transduction in glial cells. The interaction with RAGE could explain the broad ligand spectrum of formyl peptide receptors and their important role for inflammation and the host defence against infections.
International Journal of Immunopathology and Pharmacology | 2009
Henrik Wilms; Philip Rosenstiel; Marina Romero-Ramos; Alexander Arlt; Dirk Seegert; Kahle Pj; Odoy S; Claasen Jh; Holzknecht C; Lars-Ove Brandenburg; G. Deuschl; Stefan Schreiber; Deniz Kirik; Ralph Lucius
α-Synuclein (α-Syn) accounts, as a major component of Lewy bodies (LB), for the filamentous deposits in many cases of neurodegenerative diseases. Yet, little is known about the molecular mechanisms of neuronal loss in these diseases. The correlation between α-Syn oligomerization/aggregation and pathologies raises the key question of which molecular form of α-Syn (i.e. monomeric α-Syn, protofibrils or mature fibrils) represents the damage-inducing culprit in the scenario of synucleinopathies. We show that human α-Syn protofibrils (PFs) are potent activators of parallel proinflammatory signalling pathways (p38 and ERK1/2 MAP kinases and NF-κB) in microglial cells in vitro. Furthermore, stereotactic injection of α-Syn PFs into the substantia nigra of adult rats leads to a profound activation of microglia and adjacent neuronal cell loss, which can be attenuated by the MAP kinase inhibitor semapimod. We propose that the neurodegenerative process of α-synucleinopathies involves microglial activation through α-Syn released or extruded from cells with pathogenic α-Syn metabolism. Compounds that inhibit the MAPK/NF-κB pathways might be a promising pharmacological strategy for the treatment of the inflammatory component of synucleinopathies including PD.
Histochemistry and Cell Biology | 2009
Deike Varoga; Christoph Jan Wruck; Mersedeh Tohidnezhad; Lars-Ove Brandenburg; Friedrich Paulsen; Rolf Mentlein; Andreas Seekamp; L. Besch; Thomas Pufe
Gram-positive bacterial bone infections are an important cause of morbidity particularly in immunocompromised patients. Antimicrobial peptides (AP) are effectors of the innate immune system and directly kill microorganisms in the first hours after microbial infection. The aim of the present investigation was to study the expression and regulation of gram-positive specialized human β-defensin-3 (HBD-3) in bone. Samples of healthy and osteomyelitic human bone were assessed for the expression of HBD-3. Using primary and immortalized osteoblasts (SAOS-2 cells), release and regulation of HBD-3 was evaluated after exposure to Staphylococcus aureus supernatant and/or corticosteroids using PCR, immunohistochemistry, Western blot and ELISA. To determine the role of toll-like-receptors-2 and -4 (TLR-2/-4), shRNA was used to downregulate TLRs. An osteomyelitis mouse model was created performed to investigate the release of murine β-defensins using immunohistochemistry and RT-PCR. Cultured osteoblasts and human bone produce HBD-3 under standard conditions. The release increases within hours of bacterial supernatant exposure in cultured osteoblasts. This observation was not made in chronically infected bone samples. The shRNA-technology revealed the necessity of TLR-2 and -4 in HBD-3 induction in osteoblasts. Blocking protein synthesis with cycloheximide showed that the rapid release of HBD-3 is not dependent on a translational de novo synthesis and is not affected by glucocorticoids. The murine osteomyelitis model confirmed the in vivo release uptake of mouse β-defensins-4 (MBD-4) in bone. This report shows the bacterial induction of HBD-3 via TLR-2 and -4 in osteoblasts and suggests a central role of antimicrobial peptides in the prevention of bacterial bone infection. The rapid and effective induction of HBD-3 in osteoblasts incubated with conditioned media from bacteria is more likely a result of a rapid secretion of preformed HBD-3 by osteoblasts rather than a result of enhanced biosynthesis. The increased incidence of gram-positive bacterial bone infection in patients with regular intake of glucocorticoids does not seem to be caused by a deranged HBD-3 release in osteoblasts.
Molecular Immunology | 2010
Lars-Ove Brandenburg; Sandra Jansen; Christoph Jan Wruck; Ralph Lucius; Thomas Pufe
Antimicrobial peptides are part of the innate immune system of many organ systems, yet little is known about their expression and function in the brain. The antibacterial cathelicidin rCRAMP in rats (homologue of the human LL-37) not only exhibits potent bactericidal activities but also functions as a chemoattractant for immune cells. In this study, to further evaluate the role of rCRAMP in innate immunity of brain cells, we investigated the impact of rCRAMP on glial cell activation. To this end we analyzed the activation of rCRAMP-induced signalling by cytokine expression, Western blotting of certain signal transduction pathways and by cAMP level measurement in primary rat glial cells (astrocytes and microglia). We demonstrate (i) the induction of proinflammatory cytokine and neurotrophic factors and (ii) the activation of various signal transduction pathways by rCRAMP in glial cells. Moreover, (iii) we have been able to show that rCRAMP-induced IL-6 expression and ERK1/2 phosphorylation in glial cells were attenuated by the antagonists for purinergic receptor P2Y, whereas P2X and FPRL1 antagonists do not show any effects. Our results indicate for the first time that a newly identified P2Y11 receptor participates in rCRAMP-induced signal transduction. This study provides evidence that rCRAMP participates in brain immunity by stimulating cytokine production and glial cell activation, and aid in the protection of brain cells by inducing neurotrophic factors.