Network


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

Hotspot


Dive into the research topics where Christoph R.W. Kuhlmann is active.

Publication


Featured researches published by Christoph R.W. Kuhlmann.


The FASEB Journal | 2010

Cellular mechanisms of IL-17-induced blood-brain barrier disruption

Jula Huppert; Dorothea Closhen; Andrew L. Croxford; Robin E. White; Paulina Kulig; Eweline Pietrowski; Ingo Bechmann; Burkhard Becher; Heiko J. Luhmann; Ari Waisman; Christoph R.W. Kuhlmann

Recently T‐helper 17 (Th17) cells were demonstrated to disrupt the blood‐brain barrier (BBB) by the action of IL‐17A. The aim of the present study was to examine the mechanisms that underlie IL‐17A‐induced BBB breakdown. Barrier integrity was analyzed in the murine brain endothelial cell line bEnd.3 by measuring the electrical resistance values using electrical call impedance sensing technology. Furthermore, in‐cell Western blots, fluorescence imaging, and mono‐cyte adhesion and transendothelial migration assays were performed. Experimental autoimmune encephzalomyelitis (EAE) was induced in C57BL/6 mice. IL‐17A induced NADPH oxidase‐ or xanthine oxidase‐dependent reactive oxygen species (ROS) production. The resulting oxidative stress activated the endothelial contractile machinery, which was accompanied by a down‐regulation of the tight junction molecule occludin. Blocking either ROS formation or myosin light chain phosphorylation or applying IL‐17A‐neutralizing antibodies prevented IL‐17A‐induced BBB disruption. Treatment of mice with EAE using ML‐7, an inhibitor of the myosin light chain kinase, resulted in less BBB disruption at the spinal cord and less infiltration of lymphocytes via the BBB and subsequently reduced the clinical characteristics of EAE. These observations indicate that IL‐17A accounts for a crucial step in the development of EAE by impairing the integrity of the BBB, involving augmented production of ROS.—Huppert, J., Closhen, D., Croxford, A., White, R., Kulig, P., Pietrowski, E., Bechmann, I., Becher, B., Luhmann, H. J., Waisman, A., Kuhlmann, C. R. W. Cellular mechanisms of IL‐17‐induced blood‐brain barrier disruption. FASEB J. 24, 1023–1034 (2010). www.fasebj.org


Journal of Biological Chemistry | 2008

The functional role of the second NPXY motif of the LRP1 beta-chain in tissue-type plasminogen activator-mediated activation of N-methyl-D-aspartate receptors.

Anne M. Martin; Christoph R.W. Kuhlmann; Svenja V. Trossbach; Sebastian Jaeger; Elaine Waldron; Anton Roebroek; Heiko J. Luhmann; Alexander Laatsch; Sascha Weggen; Volkmar Lessmann; Claus U. Pietrzik

The low density lipoprotein receptor-related protein 1 (LRP1) emerges to play fundamental roles in cellular signaling pathways in the brain. One of its prominent ligands is the serine proteinase tissue-type plasminogen activator (tPA), which has been shown to act as a key activator of neuronal mitogen-activated protein kinase pathways via the N-methyl-d-aspartate (NMDA) receptor. However, here we set out to examine whether LRP1 and the NMDA receptor might eventually act in a combined fashion to mediate tPA downstream signaling. By blocking tPA from binding to LRP1 using the receptor-associated protein, we were able to completely inhibit NMDA receptor activation. Additionally, inhibition of NMDA receptor calcium influx with MK-801 resulted in dramatic reduction of tPA-mediated downstream signaling. This indicates a functional interaction between the two receptors, since both experimental approaches resulted in strongly reduced calcium influx and Erk1/2 phosphorylation. Additionally, we were able to inhibit Erk1/2 activation by competing for the LRP1 C-terminal binding motif with a truncated PSD95 construct resembling its PDZ III domain. Furthermore, we identified the distal NPXY amino acid motif in the C terminus of LRP1 as the crucial element for LRP1-NMDA receptor interaction via the adaptor protein PSD95. These results provide new insights into the mechanism of a tPA-induced, LRP1-mediated gating mechanism for NMDA receptors.


Journal of Investigative Dermatology | 2013

An Alternative Pathway of Imiquimod-Induced Psoriasis-Like Skin Inflammation in the Absence of Interleukin-17 Receptor A Signaling

Khalifa El Malki; Susanne Karbach; Jula Huppert; Morad Zayoud; Sonja Reißig; Rebecca Schüler; Alexej Nikolaev; Khalad Karram; Thomas Münzel; Christoph R.W. Kuhlmann; Heiko J. Luhmann; Esther von Stebut; Simone Wörtge; Florian C. Kurschus; Ari Waisman

Topical application of imiquimod (IMQ) on the skin of mice induces inflammation with common features found in psoriatic skin. Recently, it was postulated that IL-17 has an important role both in psoriasis and in the IMQ model. To further investigate the impact of IL-17RA signaling in psoriasis, we generated IL-17 receptor A (IL-17RA)-deficient mice (IL-17RA(del)) and challenged these mice with IMQ. Interestingly, the disease was only partially reduced and delayed but not abolished when compared with controls. In the absence of IL-17RA, we found persisting signs of inflammation such as neutrophil and macrophage infiltration within the skin. Surprisingly, already in the naive state, the skin of IL-17RA(del) mice contained significantly elevated numbers of Th17- and IL-17-producing γδ T cells, assuming that IL-17RA signaling regulates the population size of Th17 and γδ T cells. Upon IMQ treatment of IL-17RA(del) mice, these cells secreted elevated amounts of tumor necrosis factor-α, IL-6, and IL-22, accompanied by increased levels of the chemokine CXCL2, suggesting an alternative pathway of neutrophil and macrophage skin infiltration. Hence, our findings have major implications in the potential long-term treatment of psoriasis by IL-17-targeting drugs.


Journal of Neurochemistry | 2007

Inhibition of the myosin light chain kinase prevents hypoxia-induced blood–brain barrier disruption

Christoph R.W. Kuhlmann; Ryo Tamaki; Martin Gamerdinger; Volkmar Lessmann; Christian Behl; Oliver Kempski; Heiko J. Luhmann

Increased mortality after stroke is associated with development of brain edema. The aim of the present study was to examine the contribution of endothelial myosin light chain (MLC) phosphorylation to hypoxia‐induced blood–brain barrier (BBB) opening. Measurements of trans‐endothelial electrical resistance (TEER) were performed to analyse BBB integrity in an in vitro co‐culture model (bovine brain microvascular endothelial cells (BEC) and rat astrocytes). Brain fluid content was analysed in rats after stroke induction using a two‐vein occlusion model. Dihydroethidium was used to monitor intracellular generation of reactive oxygen species (ROS) in BEC. MLC phosphorylation was detected using immunohistochemistry and immunoblot analysis. Hypoxia caused a decrease of TEER values by more than 40%, which was prevented by inhibition of the MLC‐kinase (ML‐7, 10 μmol/L). In addition, ML‐7 significantly reduced the brain fluid content in vivo after stroke. The NAD(P)H‐oxidase inhibitor apocynin (500 μmol/L) prevented the hypoxia‐induced TEER decrease. Hypoxia‐dependent ROS generation was completely abolished by apocynin. Furthermore, ML‐7 and apocynin blocked hypoxia‐dependent phosphorylation of MLC. Our data demonstrate that hypoxia causes a breakdown of the BBB in vitro and in vivo involving ROS and the contractile machinery.


Free Radical Biology and Medicine | 2009

Oxidative stress upregulates the NMDA receptor on cerebrovascular endothelium.

Christian Betzen; Robin White; Christoph M. Zehendner; Eweline Pietrowski; Bianca Bender; Heiko J. Luhmann; Christoph R.W. Kuhlmann

N-methyl-d-aspartate receptor (NMDA-R)-mediated oxidative stress has been implicated in blood-brain barrier (BBB) disruption in a variety of neuropathological diseases. Although some interactions between both phenomena have been elucidated, possible influences of reactive oxygen species (ROS) on the NMDA-R itself have so far been neglected. The objective of this study was to examine how the cerebroendothelial NMDA-R is affected by exposure to oxidative stress and to assess possible influences on BBB integrity. RT-PCR confirmed several NMDA-R subunits (NR1, NR2B-D) expressed in the bEnd3 cell line (murine cerebrovascular endothelial cells). NR1 protein expression after exposure to ROS was observed via in-cell Western. The functionality of the expressed NMDA-R was determined by measuring DiBAC fluorescence in ROS-preexposed cells upon stimulation with the specific agonist NMDA. Finally, the effects on barrier integrity were evaluated using the ECIS system to detect changes in monolayer impedance upon NMDA-R stimulation after exposure to ROS. The expression of NR1 significantly (p<0.001) increased 72 h after 30 min exposure to superoxide (+33.8+/-7.5%), peroxynitrite (+84.9+/-10.7%), or hydrogen peroxide (+92.8+/-7.6%), resulting in increased cellular response to NMDA-R stimulation and diminished monolayer impedance. We conclude that oxidative stress upregulates NMDA-R on cerebrovascular endothelium and thus heightens susceptibility to glutamate-induced BBB disruption.


Biomacromolecules | 2008

Novel Fluorescent Core-Shell Nanocontainers for Cell Membrane Transport

Meizhen Yin; Christoph R.W. Kuhlmann; Ksenia Sorokina; Chen Li; George Mihov; Eweline Pietrowski; Kaloian Koynov; Markus Klapper; Heiko J. Luhmann; Klaus Müllen; Tanja Weil

The synthesis and characterization of novel core-shell macromolecules consisting of a fluorescent perylene-3,4,9,10-tetracarboxdiimide chromophore in the center surrounded by a hydrophobic polyphenylene shell as a first and a flexible hydrophilic polymer shell as a second layer was presented. Following this strategy, several macromolecules bearing varying polymer chain lengths, different polymer shell densities, and increasing numbers of positive and negative charges were achieved. Because all of these macromolecules reveal a good water solubility, their ability to cross cellular membranes was investigated. In this way, a qualitative relationship between the molecular architecture of these macromolecules and the biological response was established.


PLOS ONE | 2011

Caspase-3 Contributes to ZO-1 and Cl-5 Tight-Junction Disruption in Rapid Anoxic Neurovascular Unit Damage

Christoph M. Zehendner; Laura Librizzi; Marco de Curtis; Christoph R.W. Kuhlmann; Heiko J. Luhmann

Background Tight-junction (TJ) protein degradation is a decisive step in hypoxic blood-brain barrier (BBB) breakdown in stroke. In this study we elucidated the impact of acute cerebral ischemia on TJ protein arrangement and the role of the apoptotic effector protease caspase-3 in this context. Methodology/Principal Findings We used an in vitro model of the neurovascular unit and the guinea pig whole brain preparation to analyze with immunohistochemical methods the BBB properties and neurovascular integrity. In both methodological approaches we observed rapid TJ protein disruptions after 30 min of oxygen and glucose deprivation or middle cerebral artery occlusion, which were accompanied by strong caspase-3 activation in brain endothelial cells (BEC). Surprisingly only few DNA-fragmentations were detected with TUNEL stainings in BEC. Z-DEVD-fmk, an irreversible caspase-3 inhibitor, partly blocked TJ disruptions and was protective on trans-endothelial electrical resistance. Conclusions/Significance Our data provide evidence that caspase-3 is rapidly activated during acute cerebral ischemia predominantly without triggering DNA-fragmentation in BEC. Further we detected fast TJ protein disruptions which could be partly blocked by caspase-3 inhibition with Z-DEVD-fmk. We suggest that the basis for clinically relevant BBB breakdown in form of TJ disruptions is initiated within minutes during ischemia and that caspase-3 contributes to this process.


Biomaterials | 2010

Fine-tuning DNA/albumin polyelectrolyte interactions to produce the efficient transfection agent cBSA-147

Klaus Eisele; Radu A. Gropeanu; Christoph M. Zehendner; Ali Rouhanipour; Arvind Ramanathan; George Mihov; Kalojan Koynov; Christoph R.W. Kuhlmann; Subhash G. Vasudevan; Heiko J. Luhmann; Tanja Weil

We present the preparation and isolation of different chemically modified BSA species with varying numbers of primary amino groups at the surface. Highly cationic albumin proteins with increased numbers of amino groups were achieved and complex formation with plasmid DNA was carefully investigated. We compare the transfection results, polyelectrolyte complexes morphologies with their impact on complex stabilities, cytotoxicities and DNA accessibility. This knowledge-driven approach led to the identification of the efficient non-viral DNA delivery agent cBSA-147, which showed high transfection efficacies and stability.


Stroke | 2009

Mechanisms of C-Reactive Protein-Induced Blood–Brain Barrier Disruption

Christoph R.W. Kuhlmann; Laura Librizzi; Dorothea Closhen; Thorsten Pflanzner; Volkmar Lessmann; Claus U. Pietrzik; Marco de Curtis; Heiko J. Luhmann

Background and Purpose— Increased mortality after stroke is associated with brain edema formation and high plasma levels of the acute phase reactant C-reactive protein (CRP). The aim of this study was to examine whether CRP directly affects blood-brain barrier stability and to analyze the underlying signaling pathways. Methods— We used a cell coculture model of the blood-brain barrier and the guinea pig isolated whole brain preparation. Results— We could show that CRP at clinically relevant concentrations (10 to 20 &mgr;g/mL) causes a disruption of the blood-brain barrier in both approaches. The results of our study further demonstrate CRP-induced activation of surface Fc&ggr; receptors CD16/32 followed by p38-mitogen-activated protein kinase-dependent reactive oxygen species formation by the NAD(P)H-oxidase. The resulting oxidative stress increased myosin light chain kinase activity leading to an activation of the contractile machinery. Blocking myosin light chain phosphorylation prevented the CRP-induced blood-brain barrier breakdown and the disruption of tight junctions. Conclusions— Our data identify a previously unrecognized mechanism linking CRP and brain edema formation and present a signaling pathway that offers new sites of therapeutic intervention.


Life Sciences | 2008

Fluvastatin prevents glutamate-induced blood-brain-barrier disruption in vitro.

Christoph R.W. Kuhlmann; Marlis Gerigk; Bianca Bender; Dorothea Closhen; Volkmar Lessmann; Heiko J. Luhmann

Glutamate is an important excitatory amino acid in the central nervous system. Under pathological conditions glutamate levels dramatically increase. Aim of the present study was to examine whether the HMG-CoA inhibitor fluvastatin prevents glutamate-induced blood-brain-barrier (BBB) disruption. Measurements of transendothelial electrical resistance (TEER) were performed to analyze BBB integrity in an in vitro co-culture model of brain endothelial and glial cells. Myosin light chain (MLC) phosphorylation was detected by immunohistochemistry, or using the in-cell western technique. Intracellular Ca2+ and reactive oxygen species (ROS) levels were analyzed using the fluorescence dyes Ca-green or DCF. Glutamate induced a time- (1-3 h) and concentration- (0.25-1 mmol/l) dependent decrease of TEER values that was blocked by the NMDA-receptor antagonist MK801, the Ca2+ chelator BAPTA, the NAD(P)H-oxidase inhibitor apocynin and the MLC-kinase inhibitor ML-7. Furthermore we observed a concentration-dependent increase of intracellular Ca2+ and ROS after glutamate application. Glutamate caused an increase of MLC phosphorylation that was antagonized by apocynin, or BAPTA, indicating that Ca2+ and ROS signaling is involved in the activation of the contractile machinery. Fluvastatin (10-25 micromol/l) completely abolished the glutamate-induced barrier disruption and oxidative stress. The BBB-protecting effect of fluvastatin was completely lost if the cells were treated with the nitric oxide (NO) synthase inhibitor L-NMMA (300 micromol/l). In the present study we demonstrated that glutamate-induced BBB disruption involves Ca2+ signalling via NMDA receptors, which is followed by an increased ROS generation by the NAD(P)H-oxidase. This oxidative stress then activates the MLC kinase. Fluvastatin preserves barrier function in a NO-dependent way and reduces glutamate-induced oxidative stress.

Collaboration


Dive into the Christoph R.W. Kuhlmann's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Volkmar Lessmann

Otto-von-Guericke University Magdeburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge