42 results on '"Kuhlmann CR"'
Search Results
2. N-3 vs. n-6 fatty acids differentially influence calcium signalling and adhesion of inflammatory activated monocytes: impact of lipid rafts.
- Author
-
Schaefer MB, Schaefer CA, Schifferings S, Kuhlmann CR, Urban A, Benscheid U, Fischer T, Hecker M, Morty RE, Vadasz I, Herold S, Witzenrath M, Seeger W, Erdogan A, and Mayer K
- Subjects
- Antigens, CD metabolism, Calcium Signaling drug effects, Cell Adhesion drug effects, Cells, Cultured, Human Umbilical Vein Endothelial Cells drug effects, Human Umbilical Vein Endothelial Cells metabolism, Human Umbilical Vein Endothelial Cells physiology, Humans, Membrane Microdomains metabolism, Monocytes metabolism, Monocytes physiology, Tumor Necrosis Factor-alpha pharmacology, U937 Cells, Arachidonic Acid pharmacology, Docosahexaenoic Acids pharmacology, Membrane Microdomains drug effects, Monocytes drug effects
- Abstract
Background: Anti-inflammatory n-3 fatty acids (FA) like docosahexaenoic acid (DHA) opposed to the pro-inflammatory n-6 FA arachidonic acid (AA) might modulate lipid rafts within the cell membrane by differential incorporation. In inflammation, monocyte adhesion to endothelial cells is a crucial step mediated by intracellular calcium changes. We investigated whether lipid rafts mediate FA-induced modulation of adhesion and intracellular calcium., Methods: In isolated human monocytes and monocytic U937 cells we measured adhesion to human umbilical vein endothelial cells (HUVEC) using a parallel flow chamber and a static assay, adhesion molecules by FACScan, and intracellular calcium by fluorescence. Monocyte lipid rafts were isolated by ultracentrifugation and submitted to gas chromatography for FA analysis., Results: Pre-incubation with AA or DHA resulted in a predominant incorporation of the respective FA into raft compared to non-raft fraction. DHA as compared to AA significantly reduced monocyte adhesion and calcium release after stimulation with TNF-α while expression of adhesion molecules remained unchanged. Pre-treatment with a calcium chelator abolished the effect of FA on calcium and adhesion. Disruption of lipid rafts prevented FA-induced modulations., Conclusion: Incorporation of FA into lipid rafts seem to be crucial for modulation of adhesion under inflammatory conditions.
- Published
- 2016
- Full Text
- View/download PDF
3. BDNF-induced nitric oxide signals in cultured rat hippocampal neurons: time course, mechanism of generation, and effect on neurotrophin secretion.
- Author
-
Kolarow R, Kuhlmann CR, Munsch T, Zehendner C, Brigadski T, Luhmann HJ, and Lessmann V
- Abstract
BDNF and nitric oxide signaling both contribute to plasticity at glutamatergic synapses. However, the role of combined signaling of both pathways at the same synapse is largely unknown. Using NO imaging with diaminofluoresceine in cultured hippocampal neurons we analyzed the time course of neurotrophin-induced NO signals. Application of exogenous BDNF, NT-4, and NT-3 (but not NGF) induced NO signals in the soma and in proximal dendrites of hippocampal neurons that were sensitive to NO synthase activity, TrkB signaling, and intracellular calcium elevation. The effect of NO signaling on neurotrophin secretion was analyzed in BDNF-GFP, and NT-3-GFP transfected hippocampal neurons. Exogenous application of the NO donor sodium-nitroprusside markedly inhibited neurotrophin secretion. However, endogenously generated NO in response to depolarization and neurotrophin stimulation, both did not result in a negative feedback on neurotrophin secretion. These results suggest that a negative feedback of NO signaling on synaptic secretion of neurotrophins operates only at high intracellular levels of nitric oxide that are under physiological conditions not reached by depolarization or BDNF signaling.
- Published
- 2014
- Full Text
- View/download PDF
4. An alternative pathway of imiquimod-induced psoriasis-like skin inflammation in the absence of interleukin-17 receptor a signaling.
- Author
-
El Malki K, Karbach SH, Huppert J, Zayoud M, Reissig S, Schüler R, Nikolaev A, Karram K, Münzel T, Kuhlmann CR, Luhmann HJ, von Stebut E, Wörtge S, Kurschus FC, and Waisman A
- Subjects
- Adjuvants, Immunologic pharmacology, Animals, Disease Models, Animal, Female, Imiquimod, Interleukin-17 metabolism, Interleukin-6 immunology, Interleukin-6 metabolism, Interleukins immunology, Interleukins metabolism, Macrophages drug effects, Macrophages immunology, Mice, Mice, Knockout, Neutrophil Infiltration drug effects, Neutrophil Infiltration immunology, Psoriasis genetics, Receptors, Interleukin-17 genetics, Receptors, Interleukin-17 metabolism, Signal Transduction drug effects, Skin immunology, Skin metabolism, Skin pathology, Interleukin-22, Aminoquinolines pharmacology, Interleukin-17 immunology, Psoriasis chemically induced, Psoriasis immunology, Receptors, Interleukin-17 immunology, Signal Transduction immunology
- Abstract
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.
- Published
- 2013
- Full Text
- View/download PDF
5. Effects of cerivastatin on adrenergic pathways, hypertrophic growth and TGFbeta expression in adult ventricular cardiomyocytes.
- Author
-
Maxeiner H, Abdallah Y, Kuhlmann CR, Schlüter KD, and Wenzel S
- Subjects
- Animals, Cells, Cultured, Enzyme Activation drug effects, Gene Expression Regulation drug effects, Heart Failure drug therapy, Heart Failure metabolism, Heart Failure pathology, Heart Ventricles pathology, Male, Myocytes, Cardiac pathology, Proto-Oncogene Proteins c-akt metabolism, Pyridines therapeutic use, Rats, Rats, Wistar, Reactive Oxygen Species metabolism, p38 Mitogen-Activated Protein Kinases metabolism, Heart Ventricles metabolism, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, MAP Kinase Signaling System drug effects, Myocytes, Cardiac metabolism, Pyridines pharmacology, Receptors, Adrenergic, alpha metabolism, Receptors, Adrenergic, beta metabolism, Transforming Growth Factor beta biosynthesis
- Abstract
The effects of statin treatment in the setting of heart failure have already been shown. Nevertheless, there is little knowledge about its influence on adrenergic pathways in cardiomyocytes. Therefore, this study investigated the impact of cerivastatin on adrenoceptor-mediated signalling pathways in isolated adult ventricular cardiomyocytes. It focused on two endpoints: hypertrophic growth and TGFbeta expression. Cultured cardiomyocytes were used to study rac activation (analysed by its translocation into the membrane fraction), ROS formation (H(2)DCF fluorescence) and hypertrophic growth ((14)C-phenylalanine incorporation). Alpha- and beta-adrenoceptor stimulation showed significant differences regarding rac activation, ROS formation, and p38 MAP kinase activation. Both alpha- and beta-adrenoceptor stimulation induced TGFbeta expression. Upon activation of alpha-adrenergic signalling - although ROS formation was not influenced by cerivastatin - TGFbeta expression decreased. Following beta stimulation, TGFbeta expression as well as rac and p38 MAP kinase activation were reduced after pre-treatment with cerivastatin. Statin treatment did not show any influence on hypertrophic growth. In summary, this study clearly demonstrates the ability of adrenoceptor stimulation to increase TGFbeta expression. One component of the beneficial effects of statin therapy on heart failure might therefore be due to a dominant reduction and inhibition of TGFbeta, which is involved in many pathophysiological processes in cardiomyocytes., (Copyright © 2012 Elsevier GmbH. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
6. LRP1 mediates bidirectional transcytosis of amyloid-β across the blood-brain barrier.
- Author
-
Pflanzner T, Janko MC, André-Dohmen B, Reuss S, Weggen S, Roebroek AJ, Kuhlmann CR, and Pietrzik CU
- Subjects
- Animals, Cells, Cultured, Gene Knock-In Techniques, Low Density Lipoprotein Receptor-Related Protein-1, Mice, Mice, 129 Strain, Mice, Inbred C57BL, Amyloid beta-Peptides metabolism, Blood-Brain Barrier metabolism, Peptide Fragments metabolism, Receptors, LDL physiology, Transcytosis physiology, Tumor Suppressor Proteins physiology
- Abstract
According to the "amyloid hypothesis", the amyloid-β (Aβ) peptide is the toxic intermediate driving Alzheimer's disease (AD) pathogenesis. Recent evidence suggests that the low density lipoprotein receptor-related protein 1 (LRP1) transcytoses Aβ out of the brain across the blood-brain barrier (BBB). To provide genetic evidence for LRP1-mediated transcytosis of Aβ across the BBB we analyzed Aβ transcytosis across primary mouse brain capillary endothelial cells (pMBCECs) derived from wild-type and LRP1 knock-in mice. Here, we show that pMBCECs in vitro express functionally active LRP1. Moreover, we demonstrate that LRP1 mediates transcytosis of [(125)I]-Aβ(1-40) across pMBCECs in both directions, whereas no role for LRP1-mediated Aβ degradation was detected. Analysis of [(125)I]-Aβ(1-40) transport across pMBCECs generated from mice harboring a knock-in mutation in the NPxYxxL endocytosis/sorting domain of endogenous LRP1 revealed a reduced Aβ clearance from brain-to-blood and blood-to-brain compared with wild-type derived pMBCECs. Therefore, for the first time, we present genetic evidence that LRP1 modulates the pathogenic actions of soluble Aβ in the brain by clearing Aβ across the BBB., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
7. Caspase-3 contributes to ZO-1 and Cl-5 tight-junction disruption in rapid anoxic neurovascular unit damage.
- Author
-
Zehendner CM, Librizzi L, de Curtis M, Kuhlmann CR, and Luhmann HJ
- Subjects
- Animals, Blood-Brain Barrier metabolism, Capillary Permeability physiology, Caspase 3 metabolism, Cells, Cultured, Endothelial Cells metabolism, Endothelial Cells physiology, Guinea Pigs, Hypoxia, Brain metabolism, Mice, Protein Transport physiology, Tight Junctions pathology, Time Factors, Zonula Occludens-1 Protein, Blood-Brain Barrier pathology, Caspase 3 physiology, Claudins metabolism, Hypoxia, Brain pathology, Membrane Proteins metabolism, Phosphoproteins metabolism, Tight Junctions metabolism
- Abstract
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.
- Published
- 2011
- Full Text
- View/download PDF
8. Pro-inflammatory effects of interleukin-17A on vascular smooth muscle cells involve NAD(P)H- oxidase derived reactive oxygen species.
- Author
-
Pietrowski E, Bender B, Huppert J, White R, Luhmann HJ, and Kuhlmann CR
- Subjects
- Acetophenones pharmacology, Animals, Aorta, Thoracic cytology, Cell Differentiation drug effects, Cell Differentiation physiology, Cell Movement drug effects, Cell Movement physiology, Cells, Cultured, Cytokines metabolism, Enzyme Inhibitors pharmacology, Interleukin-17 pharmacology, Membrane Glycoproteins antagonists & inhibitors, Membrane Glycoproteins genetics, Mice, Mice, Inbred C57BL, Muscle, Smooth, Vascular cytology, NADPH Oxidase 2, NADPH Oxidase 4, NADPH Oxidases antagonists & inhibitors, NADPH Oxidases genetics, Oxidative Stress drug effects, Oxidative Stress physiology, RNA, Small Interfering, p38 Mitogen-Activated Protein Kinases metabolism, Interleukin-17 metabolism, Membrane Glycoproteins metabolism, Muscle, Smooth, Vascular enzymology, Muscle, Smooth, Vascular immunology, NADPH Oxidases metabolism, Reactive Oxygen Species metabolism
- Abstract
T cells are known for their contribution to the inflammatory element of atherosclerosis. Recently, it has been demonstrated that the Th17 derived cytokine IL-17 is involved in the pro-inflammatory response of vascular smooth muscle cells (VSMC). The aim of the present study was to examine whether reactive oxygen species (ROS) might be involved in this context. The effect of IL-17A on ROS generation was examined using the fluorescent dye 2'7'-dichlorodihydrofluorescein (H(2)DCF) in primary murine VSMC. IL-17A induced an increase in H(2)DCF fluorescence in VSMC, and this effect was blocked by the NAD(P)H-oxidase inhibitor apocynin and siRNA targeting Nox2. The p38-MAPK inhibitors SB203580 and SB202190 dose-dependently reduced the IL-17A induced ROS production. The IL-17A induced release of the pro-inflammatory cytokines IL-6, G-CSF, GM-CSF and MCP-1 from VSMC, as detected by the Luminex technology, was completely abolished by NAD(P)H-oxidase inhibition. Taken together, our data indicate that IL-17A causes the NAD(P)H-oxidase dependent generation of ROS leading to a pro-inflammatory activation of VSMC., (Copyright © 2010 S. Karger AG, Basel.)
- Published
- 2011
- Full Text
- View/download PDF
9. Blood-brain-barrier models for the investigation of transporter- and receptor-mediated amyloid-β clearance in Alzheimer's disease.
- Author
-
Pflanzner T, Kuhlmann CR, and Pietrzik CU
- Subjects
- Alzheimer Disease pathology, Animals, Endothelial Cells metabolism, Endothelial Cells pathology, Mice, Models, Biological, Alzheimer Disease metabolism, Amyloid beta-Peptides metabolism, Blood-Brain Barrier metabolism, Membrane Transport Proteins physiology, Receptors, Cell Surface physiology, Transcytosis physiology
- Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly with more than 26 million people worldwide living with the disease. Besides the main neuropathological hallmarks of AD, provoked by the accumulation of amyloid-β (Aβ) and tau hyperphosphorylation, other cells and cellular systems such as microglia and the neurovascular unit establishing the blood-brain-barrier (BBB) have been implicated to play a role in AD etiopathology. Insulating the brain from the blood stream, the BBB facilitates supply and disposal of nutrients and metabolites by the expression of transporters and transcytotic receptors at the polarized endothelial cell (EC) surface. Recently, several proteins involved in Aβ transport across the BBB have been identified in in vitro and in vivo studies. In this review, we summarize recent evidence of receptor- and transporter-mediated Aβ clearance across the BBB. Furthermore, we discuss the models used to identify and characterize Aβ transport across the BBB in regard to barrier properties and suitability of the models for the experimental investigation of transport mechanisms involved in Aβ clearance across an EC barrier.
- Published
- 2010
- Full Text
- View/download PDF
10. Fine-tuning DNA/albumin polyelectrolyte interactions to produce the efficient transfection agent cBSA-147.
- Author
-
Eisele K, Gropeanu RA, Zehendner CM, Rouhanipour A, Ramanathan A, Mihov G, Koynov K, Kuhlmann CR, Vasudevan SG, Luhmann HJ, and Weil T
- Subjects
- Animals, Cell Death drug effects, Clathrin metabolism, Endosomes drug effects, Endosomes metabolism, Ethidium chemistry, Green Fluorescent Proteins metabolism, Humans, Male, Middle Aged, Molecular Weight, Particle Size, Plasmids metabolism, Serum Albumin, Bovine toxicity, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Static Electricity, Sus scrofa, Thermodynamics, DNA metabolism, Electrolytes metabolism, Serum Albumin, Bovine metabolism, Transfection methods
- Abstract
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., (Copyright © 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
11. CRP-induced levels of oxidative stress are higher in brain than aortic endothelial cells.
- Author
-
Closhen D, Bender B, Luhmann HJ, and Kuhlmann CR
- Subjects
- Animals, Cattle, Endothelial Cells enzymology, NADPH Oxidases metabolism, Receptors, IgG metabolism, Aorta cytology, Brain cytology, C-Reactive Protein pharmacology, Endothelial Cells drug effects, Endothelial Cells metabolism, Oxidative Stress drug effects
- Abstract
C-reactive protein (CRP) has been demonstrated to induce blood-brain barrier disruption (BBB) involving NAD(P)H-oxidase dependent oxidative stress. It is unclear why CRP affects the BBB and not other vascular beds following stroke. Therefore we examined CRP receptor and NAD(P)H-oxidase expression levels in bovine brain- (BEC) and aortic endothelial cells. Dichlorodihydrofluorescein measurements revealed significantly higher CRP-induced reactive oxygen species (ROS) levels in BEC. Protein expression of the CRP-receptors CD16, CD32 and of the NAD(P)H-oxidase subunit p22phox were also significantly higher in BEC. In conclusion BEC show a higher vulnerability to CRP due to increased levels of CRP receptors and the NAD(P)H-oxidase., (Copyright (c) 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
12. Cellular mechanisms of IL-17-induced blood-brain barrier disruption.
- Author
-
Huppert J, Closhen D, Croxford A, White R, Kulig P, Pietrowski E, Bechmann I, Becher B, Luhmann HJ, Waisman A, and Kuhlmann CR
- Subjects
- Animals, Antibodies, Neutralizing immunology, Antibodies, Neutralizing pharmacology, Azepines pharmacology, Blood-Brain Barrier immunology, Blood-Brain Barrier pathology, Cell Line, Transformed, Down-Regulation drug effects, Down-Regulation immunology, Encephalomyelitis, Autoimmune, Experimental immunology, Encephalomyelitis, Autoimmune, Experimental pathology, Endothelial Cells immunology, Endothelial Cells pathology, Enzyme Inhibitors pharmacology, Interleukin-17 antagonists & inhibitors, Interleukin-17 immunology, Interleukin-17 pharmacology, Membrane Proteins immunology, Membrane Proteins metabolism, Mice, Mice, Inbred BALB C, Myosin-Light-Chain Kinase, NADPH Oxidases immunology, NADPH Oxidases metabolism, Naphthalenes pharmacology, Occludin, Oxidative Stress drug effects, Oxidative Stress immunology, Reactive Oxygen Species immunology, T-Lymphocytes, Helper-Inducer immunology, T-Lymphocytes, Helper-Inducer pathology, Xanthine Oxidase immunology, Xanthine Oxidase metabolism, Blood-Brain Barrier metabolism, Encephalomyelitis, Autoimmune, Experimental metabolism, Endothelial Cells metabolism, Interleukin-17 metabolism, Reactive Oxygen Species metabolism, T-Lymphocytes, Helper-Inducer metabolism
- Abstract
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 monocyte adhesion and transendothelial migration assays were performed. Experimental autoimmune encephalomyelitis (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.
- Published
- 2010
- Full Text
- View/download PDF
13. Inhibition of myosin light chain kinase reduces brain edema formation after traumatic brain injury.
- Author
-
Luh C, Kuhlmann CR, Ackermann B, Timaru-Kast R, Luhmann HJ, Behl C, Werner C, Engelhard K, and Thal SC
- Subjects
- Animals, Blood-Brain Barrier drug effects, Brain Edema etiology, Brain Edema pathology, Constriction, Disease Models, Animal, Drug Administration Schedule, Evans Blue, Functional Laterality, Gene Expression Regulation drug effects, Intracranial Pressure drug effects, Male, Mice, Mice, Inbred C57BL, Myosin Light Chains genetics, Myosin Light Chains metabolism, Myosin-Light-Chain Kinase genetics, Neurologic Examination methods, Statistics, Nonparametric, Time Factors, Azepines therapeutic use, Brain Edema drug therapy, Brain Edema metabolism, Brain Injuries complications, Enzyme Inhibitors therapeutic use, Myosin-Light-Chain Kinase metabolism, Naphthalenes therapeutic use
- Abstract
The role of the endothelial contractile apparatus in the process of brain edema formation after brain trauma is not characterized. Phosphorylation of myosin light chains by myosin light chain kinases (MLCK) activates endothelial contractile elements and results in a rearrangement of the cytoskeleton. This may enhance post-traumatic blood-brain barrier dysfunction. In order to investigate the role of the MLCK on brain edema formation and blood-brain barrier permeability after brain injury, mice were anesthetized and subjected to a controlled cortical impact (CCI). MLCK expression is significantly up-regulated after CCI with a maximum 12 h post-injury. Specific inhibition of MLCK by ML-7 resulted in a reduction of phosphorylation of myosin light chains and improved blood-brain-barrier integrity. Accordingly, ML-7 attenuated post-traumatic brain edema formation and intracranial hypertension 24 h after CCI. Prevention of brain edema formation did not translate into improved neurological outcome or reduced brain lesion. In conclusion, the results confirm that the endothelial contractile apparatus is activated by CCI and opens the endothelial barrier leading to vasogenic brain edema formation. Lack of neurological and histological improvement suggests that specific targeting of vasogenic brain edema at the endothelial level is not sufficient to limit secondary brain damage and has, therefore, to be combined with other potential neuroprotective strategies.
- Published
- 2010
- Full Text
- View/download PDF
14. Studying the neurovascular unit: an improved blood-brain barrier model.
- Author
-
Zehendner CM, Luhmann HJ, and Kuhlmann CR
- Subjects
- Animals, Brain blood supply, Caspase 3 metabolism, Cell Line, Electric Impedance, Endothelial Cells metabolism, Immunohistochemistry, Membrane Proteins analysis, Membrane Proteins metabolism, Mice, Reactive Oxygen Species analysis, Reactive Oxygen Species metabolism, Tight Junctions chemistry, Blood-Brain Barrier cytology, Blood-Brain Barrier metabolism, Brain cytology, Coculture Techniques methods, Endothelial Cells cytology, Tight Junctions metabolism
- Abstract
The blood-brain barrier (BBB) closely interacts with the neuronal parenchyma in vivo. To replicate this interdependence in vitro, we established a murine coculture model composed of brain endothelial cell (BEC) monolayers with cortical organotypic slice cultures. The morphology of cell types, expression of tight junctions, formation of reactive oxygen species, caspase-3 activity in BECs, and alterations of electrical resistance under physiologic and pathophysiological conditions were investigated. This new BBB model allows the application of techniques such as laser scanning confocal microscopy, immunohistochemistry, fluorescent live cell imaging, and electrical cell substrate impedance sensing in real time for studying the dynamics of BBB function under defined conditions.
- Published
- 2009
- Full Text
- View/download PDF
15. Oxidative stress upregulates the NMDA receptor on cerebrovascular endothelium.
- Author
-
Betzen C, White R, Zehendner CM, Pietrowski E, Bender B, Luhmann HJ, and Kuhlmann CR
- Subjects
- Animals, Apoptosis, Blood-Brain Barrier metabolism, Blotting, Western, Cells, Cultured, Cerebrovascular Circulation, Endothelium, Vascular cytology, Glutamic Acid pharmacology, Immunoenzyme Techniques, Mice, N-Methylaspartate pharmacology, RNA, Messenger genetics, RNA, Messenger metabolism, Reactive Oxygen Species metabolism, Receptors, N-Methyl-D-Aspartate genetics, Reverse Transcriptase Polymerase Chain Reaction, Endothelium, Vascular metabolism, Oxidative Stress, Receptors, N-Methyl-D-Aspartate metabolism
- Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
16. MK801 blocks hypoxic blood-brain-barrier disruption and leukocyte adhesion.
- Author
-
Kuhlmann CR, Zehendner CM, Gerigk M, Closhen D, Bender B, Friedl P, and Luhmann HJ
- Subjects
- Acetophenones pharmacology, Animals, Astrocytes, Brain cytology, Calcium metabolism, Cell Adhesion physiology, Cell Hypoxia drug effects, Cells, Cultured, Coculture Techniques, Electric Impedance, Endothelial Cells, Enzyme Inhibitors pharmacology, Glutamic Acid metabolism, Macrocyclic Compounds pharmacology, Oxazoles pharmacology, Oxygen pharmacology, Reactive Oxygen Species metabolism, Ryanodine pharmacology, Swine, Blood-Brain Barrier drug effects, Cell Adhesion drug effects, Dizocilpine Maleate pharmacology, Excitatory Amino Acid Antagonists pharmacology, Leukocytes drug effects
- Abstract
The aim of the present study was to examine the signaling pathways of hypoxia followed by reoxygenation (H/R)-induced disruption of the blood-brain-barrier (BBB) in a co-culture of astrocytes and brain endothelial cells (BEC) in vitro. We analyzed the possible stabilizing effect of MK801, a highly selective N-methyl-d-aspartate receptor (NMDAR) antagonist, on BBB integrity. Levels of reactive oxygen species (ROS), glutamate (Glut) release and monocyte adhesion were measured under normoxia and H/R. BBB integrity was monitored measuring the trans-endothelial electrical resistance (TEER). TEER values dropped under H/R conditions which was abolished by MK801. Glut release from astrocytes, but not from endothelial cells was significantly increased under H/R, as were ROS levels and monocyte adhesion. The oxidative stress was blocked by MK801 and the NAD(P)H-oxidase inhibitor apocynin. We observed that calcium (Ca(2+)) signaling plays a crucial role during ROS generation and monocyte adhesion under H/R. ROS levels were decreased by applying ryanodine, a blocker of Ca(2+) release from the endoplasmic reticulum (ER) and by lowering the extracellular Ca(2+) concentration. Xestospongin C, which blocks IP(3) mediated Ca(2+) release from the ER did not alter ROS production under H/R conditions. These findings indicate that both extracellular Ca(2+) influx and ryanodine-mediated intracellular Ca(2+) release from the ER during H/R contribute to ROS formation at the BBB. Blocking ROS or Ca(2+) signaling prevented H/R-induced monocyte adhesion to BEC. We conclude, that the activation of NMDAR under H/R by Glut increases intracellular Ca(2+) levels, contributes to BBB disruption, ROS generation and monocyte adhesion.
- Published
- 2009
- Full Text
- View/download PDF
17. Fluvastatin prevents glutamate-induced blood-brain-barrier disruption in vitro.
- Author
-
Kuhlmann CR, Gerigk M, Bender B, Closhen D, Lessmann V, and Luhmann HJ
- Subjects
- Animals, Calcium metabolism, Cell Line, Dose-Response Relationship, Drug, Electric Impedance, Endothelial Cells drug effects, Endothelial Cells metabolism, Fluvastatin, Intracellular Space drug effects, Intracellular Space metabolism, Myosin Light Chains metabolism, Nitric Oxide metabolism, Oxidative Stress drug effects, Phosphorylation drug effects, Rats, Reactive Oxygen Species metabolism, Receptors, N-Methyl-D-Aspartate metabolism, Signal Transduction drug effects, Time Factors, Blood-Brain Barrier drug effects, Blood-Brain Barrier pathology, Fatty Acids, Monounsaturated pharmacology, Glutamic Acid toxicity, Indoles pharmacology
- Abstract
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.
- Published
- 2008
- Full Text
- View/download PDF
18. Novel fluorescent core-shell nanocontainers for cell membrane transport.
- Author
-
Yin M, Kuhlmann CR, Sorokina K, Li C, Mihov G, Pietrowski E, Koynov K, Klapper M, Luhmann HJ, Müllen K, and Weil T
- Subjects
- Biological Transport, Polymers pharmacokinetics, Cell Membrane metabolism, Drug Carriers pharmacokinetics, Nanoparticles chemistry, Polymers chemistry
- Abstract
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.
- Published
- 2008
- Full Text
- View/download PDF
19. Apigenin-induced nitric oxide production involves calcium-activated potassium channels and is responsible for antiangiogenic effects.
- Author
-
Erdogan A, Most AK, Wienecke B, Fehsecke A, Leckband C, Voss R, Grebe MT, Tillmanns H, Schaefer CA, and Kuhlmann CR
- Subjects
- Cell Movement, Cells, Cultured, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, Humans, Models, Biological, Phosphorylation, Potassium Channels chemistry, Radioimmunoassay, Signal Transduction, Time Factors, Umbilical Veins metabolism, Angiogenesis Inhibitors pharmacology, Apigenin metabolism, Calcium metabolism, Nitric Oxide metabolism, Potassium Channels metabolism
- Abstract
Background: The dietary flavonoid apigenin (Api) has been demonstrated to exert multiple beneficial effects upon the vascular endothelium. The aim of this study was to examine whether Ca(2+)-activated K(+) channels (K(Ca)) are involved in endothelial nitric oxide (NO) production and antiangiogenic effects., Methods: Endothelial NO generation was monitored using a cyclic guanosine monophosphate radioimmunoassay. K(Ca) activity and changes of the intracellular Ca(2+) concentration [Ca(2+)](i) were analyzed using the fluorescent dyes bis-barbituric acid oxonol, potassium-binding benzofuran isophthalate, and fluo-3. The endothelial angiogenic parameters measured were cell proliferation, [(3)H]-thymidine incorporation, and cell migration (scratch assay). Akt phosphorylation was examined using immunohistochemistry., Results: Api caused a concentration-dependent increase in cyclic guanosine monophosphate levels, with a maximum effect at a concentration of 1 mum. Api-induced hyperpolarization was blocked by the small and large conductance K(Ca) inhibitors apamin and iberiotoxin, respectively. Furthermore, apamin and iberiotoxin blocked the late, long-lasting plateau phase of the Api-induced biphasic increase of [Ca(2+)](i). Inhibition of Ca(2+) signaling and the K(Ca) blockade both blocked NO production. Prevention of all three (NO, Ca(2+), and K(Ca) signaling) reversed the antiangiogenic effects of Api under both basal and basic fibroblast growth factor-induced culture conditions. Basic fibroblast growth factor-induced Akt phosphorylation was also reduced by Api., Conclusions: Based on our experimental results we propose the following signaling cascade for the effects of Api on endothelial cell signaling. Api activates small and large conductance K(Ca), leading to a hyperpolarization that is followed by a Ca(2+) influx. The increase of [Ca(2+)](i) is responsible for an increased NO production that mediates the antiangiogenic effects of Api via Akt dephosphorylation.
- Published
- 2007
- Full Text
- View/download PDF
20. Inhibition of the myosin light chain kinase prevents hypoxia-induced blood-brain barrier disruption.
- Author
-
Kuhlmann CR, Tamaki R, Gamerdinger M, Lessmann V, Behl C, Kempski OS, and Luhmann HJ
- Subjects
- Acetophenones pharmacology, Animals, Astrocytes drug effects, Astrocytes metabolism, Blood-Brain Barrier drug effects, Blood-Brain Barrier physiopathology, Brain Edema drug therapy, Brain Edema metabolism, Brain Edema physiopathology, Cattle, Cells, Cultured, Coculture Techniques, Electric Impedance, Endothelial Cells drug effects, Endothelial Cells metabolism, Enzyme Inhibitors pharmacology, Extracellular Fluid drug effects, Extracellular Fluid metabolism, Hypoxia, Brain drug therapy, Hypoxia, Brain physiopathology, Male, Myosin-Light-Chain Kinase antagonists & inhibitors, NADPH Oxidases antagonists & inhibitors, NADPH Oxidases metabolism, Oxidative Stress drug effects, Oxidative Stress physiology, Phosphorylation drug effects, Rats, Rats, Wistar, Reactive Oxygen Species metabolism, Vasoconstriction drug effects, Vasoconstriction physiology, Azepines pharmacology, Blood-Brain Barrier enzymology, Hypoxia, Brain enzymology, Myosin Light Chains metabolism, Myosin-Light-Chain Kinase metabolism, Naphthalenes pharmacology
- Abstract
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 micromol/L). In addition, ML-7 significantly reduced the brain fluid content in vivo after stroke. The NAD(P)H-oxidase inhibitor apocynin (500 micromol/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.
- Published
- 2007
- Full Text
- View/download PDF
21. Sildenafil inhibits the proliferation of cultured human endothelial cells.
- Author
-
Erdogan A, Luedders DW, Muenz BM, Schaefer CA, Tillmanns H, Wiecha J, and Kuhlmann CR
- Abstract
The proliferation of endothelial cells plays a crucial role in the development of intraplaque angiogenesis (IPA). IPA is a major source of intraplaque hemorrhage and therefore contributes to the destabilization of atherosclerotic plaques. Therefore, the aim of the present study was to examine, whether sildenafil inhibits endothelial cell growth. The proliferation of human endothelial cells derived from umbilical cord veins (HUVEC) was examined on DNA level by measurements of ((3)H)-thymidine incorporation. Cell viability was analyzed using trypan blue staining. The proliferation of cultured human endothelial cells was significantly decreased by 1 μmol/l (-48.4%) and 10 μmol/l (-89.6%) sildenafil (n=10, p<0.05). This was not a cytotoxic effect, because cell viability was only reduced at sildenafil concentrations of 50 μmol/l or greater. In addition sildenafil significantly reduced endothelial proliferation induced by bFGF (n=10, p<0.05). The presented results demonstrate an antiangiogenic effect of sildenafil that might be useful in the prevention of atherosclerotic plaque vascularization.
- Published
- 2007
22. The role of poly(ADP-ribose) polymerase (PARP) in the autonomous proliferative response of endothelial cells to hypoxia.
- Author
-
Abdallah Y, Gligorievski D, Kasseckert SA, Dieterich L, Schäfer M, Kuhlmann CR, Noll T, Sauer H, Piper HM, and Schäfer C
- Subjects
- Animals, Butadienes pharmacology, Calcium analysis, Calcium metabolism, Cell Hypoxia physiology, Cell Proliferation drug effects, Cells, Cultured, Cytosol chemistry, Cytosol metabolism, Endothelial Cells cytology, Enzyme Activation, Extracellular Signal-Regulated MAP Kinases analysis, Extracellular Signal-Regulated MAP Kinases metabolism, Hydrogen Peroxide metabolism, Immunohistochemistry, Microscopy, Fluorescence, NADPH Oxidases genetics, Nitriles pharmacology, Oligonucleotides, Antisense pharmacology, Phenanthrenes pharmacology, Poly(ADP-ribose) Polymerase Inhibitors, Reactive Oxygen Species metabolism, Swine, Endothelial Cells metabolism, Endothelium, Vascular, MAP Kinase Signaling System, Poly(ADP-ribose) Polymerases physiology
- Abstract
Objective: The autonomous proliferative response of endothelial cells to hypoxia has been shown to be dependent on activation of NAD(P)H oxidase, on the cytosolic Ca2+ load, and, consequently, on nuclear translocation of extracellular signal-regulated kinase (ERK)1/2 during transient hypoxia. The aim of the present study was to investigate whether poly(ADP-ribose) polymerase (PARP) is a downstream signal of NAD(P)H oxidase, mediating cytosolic Ca2+ load and hence nuclear translocation of ERK1/2 and endothelial cell proliferation., Methods: Porcine aortic endothelial cells were incubated under hypoxic conditions for 40 min. Cytosolic [Ca2+] and reactive oxygen species (ROS) formation were measured in fura-2- and DCF-loaded cells, respectively. PARP activation was detected by immunocytochemistry, and endothelial cell proliferation was determined 24 h after 60 min of transient hypoxia., Results: Inhibition of NAD(P)H oxidase with antisense oligonucleotide against the p22(phox) subunit, MEK/ERK signalling with UO 126 (30 microM), or PARP with PJ 34 (10 microM) leads to a marked reduction in hypoxia-induced cytosolic Ca2+ load and activation of PARP. Hypoxia-induced translocation of ERK1/2 and endothelial cell proliferation were also prevented when NAD(P)H oxidase or PARP were inhibited; however, hypoxic ROS formation was not affected in the presence of PARP inhibitor., Conclusion: PARP represents a downstream effector of NADP(H) oxidase and acts as a necessary intermediate step for the hypoxic proliferative response of endothelial cells.
- Published
- 2007
- Full Text
- View/download PDF
23. Activation of Ca2+ -activated potassium channels is involved in lysophosphatidylcholine-induced monocyte adhesion to endothelial cells.
- Author
-
Erdogan A, Schaefer MB, Kuhlmann CR, Most A, Hartmann M, Mayer K, Renner FC, Schaefer C, Abdallah Y, Hoelschermann H, and Schaefer CA
- Subjects
- Atherosclerosis metabolism, Calcium metabolism, Cell Adhesion drug effects, Endothelial Cells cytology, Endothelial Cells drug effects, Endothelial Cells immunology, Enzyme Inhibitors pharmacology, Humans, Large-Conductance Calcium-Activated Potassium Channels antagonists & inhibitors, Lysophosphatidylcholines pharmacology, NADPH Oxidases antagonists & inhibitors, NADPH Oxidases metabolism, Onium Compounds pharmacology, Peptides pharmacology, Reactive Oxygen Species metabolism, U937 Cells, Umbilical Veins cytology, Vasculitis metabolism, Atherosclerosis immunology, Cell Adhesion immunology, Endothelium, Vascular cytology, Large-Conductance Calcium-Activated Potassium Channels physiology, Monocytes cytology, Vasculitis immunology
- Abstract
Objective: Ca(2+)-activated K(+)-channels (BK(Ca)) play an important role in lysophosphatidylcholine (LPC)-induced endothelial dysfunction. Aim of our study was to investigate whether LPC-induced activation of BK(Ca) is also involved in monocyte adhesion to endothelial cells (EC)., Methods and Results: Measurement of membrane potential (MP) was performed using the fluorescence dye DiBAC. Adhesion of the monocytotic cell line U937 to EC was analysed by (3)[H]-thymidine-adhesion-assay. Expression of ICAM-1 and VCAM-1 were analyzed by FACS. LPC induced a hyperpolarization of EC in a dose-dependent manner with the maximum seen with 2 microM. This was prevented by the BK(Ca)-inhibitor iberiotoxin (IBX, 100nM). Adhesion of U937 cells to EC was increased after stimulation of EC with LPC. This effect was time-dependent with the maximum seen after 4h. LPC-induced adhesion was significantly reduced when EC were co-incubated with IBX, or NAD(P)H oxidase inhibitor diphenyleneiodonium (DPI, 5 microM) and also blocked by addition of 2-aminoethoxydiphenylborate (2-APB, 100 microM) or the calcium-chelator BAPTA (10 microM). Stimulation of U937 cells with LPC did not result in an increased adhesion to unstimulated EC., Conclusion: Activation of the endothelial BK(Ca) plays an important role in monocyte adhesion to endothelial cells.
- Published
- 2007
- Full Text
- View/download PDF
24. Fluvastatin stabilizes the blood-brain barrier in vitro by nitric oxide-dependent dephosphorylation of myosin light chains.
- Author
-
Kuhlmann CR, Lessmann V, and Luhmann HJ
- Subjects
- Analysis of Variance, Animals, Astrocytes drug effects, Astrocytes physiology, Biological Transport drug effects, Cattle, Cells, Cultured, Coculture Techniques methods, Dose-Response Relationship, Drug, Drug Interactions, Electric Impedance, Endothelial Cells drug effects, Endothelial Cells physiology, Enzyme Inhibitors pharmacology, Fluvastatin, Mice, Microscopy, Confocal methods, Models, Animal, Rats, omega-N-Methylarginine pharmacology, Antioxidants pharmacology, Blood-Brain Barrier drug effects, Capillary Permeability drug effects, Fatty Acids, Monounsaturated pharmacology, Indoles pharmacology, Myosin Light Chains metabolism, Nitric Oxide metabolism
- Abstract
Inhibition of the 3-hydroxy-3-methylglutaryl-coenzyme-A reductase and the downstream mevalonate pathway is in part responsible for the beneficial effects that statins exert on the cardiovascular system. In this study we aimed at analysing the stabilizing effects of fluvastatin on the blood-brain barrier (BBB) integrity, using an in vitro co-culture model of ECV304 and C6, or primary bovine endothelial cells and rat astrocytes. Fluvastatin dose-dependently (1-25 micromol/l) increased barrier integrity as analysed by measurements of transendothelial electrical resistance (TEER). This effect (117.4+/-2.6% at 25 micromol/l) was significantly reduced by the nitric oxide (NO) synthase inhibitor L-NMMA (300 micromol/l; P<0.01, n=4). The fluvastatin-induced increase of intracellular NO, as analysed by confocal DAF-fluorescence imaging, and the increase in TEER values were significantly reduced in the presence of the isoprenoid geranylgeranyl pyrophosphate (GGPP; 10 micromol/l), whereas farnesyl pyrophosphate had no effect. Immunofluorescent detection of phosphorylated myosin light chains (MLC) revealed a fluvastatin-induced (25 micromol/l) significant reduction of MLC phosphorylation (85.4+/-2.7% control, P<0.001, n=20). This effect was absent if the MLC phosphatase was blocked by calyculin (10 nmol/l). In conclusion, our data demonstrate a BBB stabilizing effect of fluvastatin that correlates with the NO-dependent dephosphorylation of endothelial MLC via the MLC phosphatase.
- Published
- 2006
- Full Text
- View/download PDF
25. Statins inhibit hypoxia-induced endothelial proliferation by preventing calcium-induced ROS formation.
- Author
-
Schaefer CA, Kuhlmann CR, Weiterer S, Fehsecke A, Abdallah Y, Schaefer C, Schaefer MB, Mayer K, Tillmanns H, and Erdogan A
- Subjects
- Acetylcysteine pharmacology, Cell Hypoxia drug effects, Cell Survival, Cells, Cultured, Chelating Agents pharmacology, Egtazic Acid analogs & derivatives, Egtazic Acid pharmacology, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, Free Radical Scavengers pharmacology, Humans, Nitric Oxide Synthase antagonists & inhibitors, Onium Compounds pharmacology, Sodium Cyanide pharmacology, Calcium metabolism, Cell Proliferation drug effects, Endothelium, Vascular drug effects, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Reactive Oxygen Species metabolism
- Abstract
Pathological hypoxia plays an important role in many diseases, such as atherosclerosis, cancer, and rheumatoid arthritis. The aim of the present study was to examine the effects of different statins on hypoxia-induced endothelial cell signalling. Human umbilical cord vein endothelial cells (HUVEC) were treated with NaCN (CN, 2.5 mmol/l) to simulate a transient hypoxia. The CN-induced increase of endothelial cell numbers was significantly (n = 10, p < 0.01) reduced by the Ca(2+) chelator BAPTA (10 micromol/l), or the reactive oxygen species (ROS) scavenger N-acetylcysteine (ACC, 1 mmol/l), or the NAD(P)H-oxidase inhibitor diphenyleneiodonium (DPI, 5 micromol/l). In detail, cell numbers were (in percentage of control): 163.24 (CN), 90.06 (CN+ACC), 92.06 (CN+DPI). Intracellular-Ca(2+) and -ROS, analysed by fluorescence imaging, were significantly increased by CN. Interestingly, the CN-induced increase of ROS was in part Ca(2+)-dependent, whereas the Ca(2+) increase was not ROS-dependent. Simvastatin (5 micromol/l), fluvastatin (2.5 micromol/l), and cerivastatin (0.1 micromol/l) all reduced CN-induced proliferation, ROS generation and Ca(2+) increase. Cell viability was not reduced by the statins and the antiproliferative effect was completely reversed by mevalonate (500 micromol/l). In conclusion our study demonstrates that statins block hypoxia-associated endothelial proliferation by preventing the increase of Ca(2+) and ROS.
- Published
- 2006
- Full Text
- View/download PDF
26. Role of cGMP in sildenafil-induced activation of endothelial Ca2+-activated K+ channels.
- Author
-
Luedders DW, Muenz BM, Li F, Rueckleben S, Tillmanns H, Waldecker B, Wiecha J, Erdogan A, Schaefer CA, and Kuhlmann CR
- Subjects
- Calcium metabolism, Cells, Cultured, Cyclic GMP-Dependent Protein Kinases physiology, Cyclic Nucleotide Phosphodiesterases, Type 5, Endothelial Cells metabolism, Humans, Membrane Potentials drug effects, Purines, Sildenafil Citrate, Sulfones, 3',5'-Cyclic-GMP Phosphodiesterases antagonists & inhibitors, Cyclic GMP physiology, Endothelial Cells drug effects, Phosphodiesterase Inhibitors pharmacology, Piperazines pharmacology, Potassium Channels, Calcium-Activated drug effects
- Abstract
Intracellular cGMP is an important second messenger in endothelial cells. Because Ca(2+)-activated K(+) channels with large conductance (BK(Ca)) have been shown to regulate endothelial cell functions, the aim of the present study was to examine whether sildenafil modulates BK(Ca) activity in cultured human endothelial cells. Changes of the endothelial cell membrane potential were analyzed using the fluorescence dye DiBAC. The patch-clamp technique was used to study BK(Ca) in human endothelial cells of umbilical cord veins (HUVEC). Intracellular Ca(2+) levels were analyzed using Fura-2 fluorescence imaging. Sildenafil caused a dose-dependent (0.05-5 micromol/l) hyperpolarization of the endothelial cells with a maximum at a concentration of 1 micromol/l. A significant increase of BK(Ca) activity was induced by sildenafil (1 micromol/l) perfusion. BK(Ca) open state-probability (NPo) was also increased by the cGMP-analogue 8-bromo-cGMP (0.5 mmol/l), whereas inhibition of the cGMP-dependent kinase (PKG) had no effect on NPo. PKG-inhibition abolished 8-bromo-cGMP induced BK(Ca) activation, and reduced sildenafil induced NPo. Furthermore, sildenafil caused a significant increase of intracellular calcium that was blocked by the BK(Ca) inhibitor iberiotoxin (100 nmol/l). In conclusion sildenafil activates BK(Ca) by a mechanism, which involves cGMP. The activation of the BK(Ca) is responsible for the sildenafil-induced increase of intracellular Ca(2+).
- Published
- 2006
- Full Text
- View/download PDF
27. Lipopolysaccharide-induced proliferation and adhesion of U937 cells to endothelial cells involves barium chloride sensitive hyperpolarization.
- Author
-
Erdogan A, Schaefer CA, Most AK, Schaefer MB, Mayer K, Tillmanns H, and Kuhlmann CR
- Subjects
- Cell Line, Tumor, Dose-Response Relationship, Drug, Drug Combinations, Endothelium, Vascular physiology, Humans, Monocytes physiology, U937 Cells, Umbilical Cord cytology, Barium Compounds pharmacology, Cell Adhesion drug effects, Cell Proliferation drug effects, Chlorides pharmacology, Endothelium, Vascular drug effects, Lipopolysaccharides pharmacology, Monocytes drug effects, Potassium Channels, Calcium-Activated drug effects
- Abstract
The adhesion of monocytes to the endothelium and their proliferation in the subendothelial space play an important role in atherosclerosis. Since the proliferation and migration of cells are influenced by the activity of ion channels, the aim of this study was to examine whether barium chloride (Ba(2+))-sensitive potassium channels (K(iCa)) are involved in lipopolysaccharide (LPS)-induced proliferation of monocytic U937 cells, and in the adhesion of these cells to endothelial cells. The adhesion of LPS-stimulated U937 cells to endothelial cells reached a maximum at a concentration of 5 microg/ml. This effect of LPS was completely abolished in the presence of Ba(2+) (100 micromol/l). In addition, LPS-induced proliferation was significantly reduced by Ba(2+) (control, 100%; LPS 5 microg/ml, 175%; LPS + Ba(2+) 100 micromol/l, 136%; n = 12, P < 0.05). To examine whether K(iCa) are activated by LPS, changes of U937 membrane potential were determined. LPS (5 microg/ml) caused a hyperpolarization of U937 cells indicating a flux of K(+) ions out of the cells. This effect was completely blocked by Ba(2+) (100 micromol/l). In conclusion, we demonstrate that LPS activates K(iCa) in U937 cells, which is responsible for LPS-induced adhesion of these cells to endothelial cells, and to the proliferation of U937 cells.
- Published
- 2006
- Full Text
- View/download PDF
28. Nicotine inhibits large conductance Ca(2+)-activated K(+) channels and the NO/-cGMP signaling pathway in cultured human endothelial cells.
- Author
-
Kuhlmann CR, Trümper JR, Tillmanns H, Alexander Schaefer C, and Erdogan A
- Subjects
- Calcium Channels physiology, Electrophysiology, Endothelial Cells physiology, Endothelins physiology, In Vitro Techniques, Nicotine adverse effects, Potassium Channels physiology, Calcium Channels drug effects, Cyclic GMP, Endothelial Cells drug effects, Endothelins drug effects, Nicotine pharmacology, Nitric Oxide, Potassium Channels drug effects, Signal Transduction drug effects
- Abstract
Objective: The effects of nicotine on endothelium-dependent vasorelaxation mediated by nitric oxide (NO) are controversial. Since endothelial NO synthesis has been shown to depend on the activity of large conductance Ca(2 + )-activated K(+) channels (BK(Ca)), the present study investigated whether nicotine alters BK(Ca) single channel activity induced by the K(+) channel opener NS1619, and to examine a possible interaction with the endothelial NO generation., Design: The patch-clamp technique was used to examine the BK(Ca) activity. NO production was measured indirectly using a [(3)H]-cGMP-radioimmunoassay. All experiments were performed using cultured endothelial cells derived from human umbilical cord veins., Results: The BK(Ca) opener NS1619 (10 micromol/l) significantly increased the BK(Ca) open-state probability (NPo) from 0.011+/-0.007 (control) to 0.052+/-0.019. Co-perfusion with nicotine (1 micromol/l) significantly decreased NS1619 induced NPo (n = 14, p < 0.05). Intracellular cGMP levels were significantly increased, if cells were stimulated with NS1619 (+ 225%; n = 10, p < 0.05), which was blocked by Nicotine (1 micromol/l)., Conclusions: The results of the present study demonstrate that BK(Ca) activation by NS1619 plays an important role in the regulation of the NO-/cGMP-signaling-pathway. Endothelial dysfunction caused by nicotine may be connected with a decrease in BK(Ca)-activity.
- Published
- 2005
- Full Text
- View/download PDF
29. Signalling mechanisms of SDF-induced endothelial cell proliferation and migration.
- Author
-
Kuhlmann CR, Schaefer CA, Reinhold L, Tillmanns H, and Erdogan A
- Subjects
- Cell Movement physiology, Cell Proliferation drug effects, Cells, Cultured, Chemokine CXCL12, Endothelial Cells drug effects, Humans, Ion Channel Gating drug effects, Large-Conductance Calcium-Activated Potassium Channels, Potassium Channels, Calcium-Activated drug effects, Signal Transduction drug effects, Cell Movement drug effects, Chemokines, CXC pharmacology, Endothelial Cells physiology, Ion Channel Gating physiology, Nitric Oxide metabolism, Potassium Channels, Calcium-Activated metabolism, Signal Transduction physiology
- Abstract
The aim of our study was to investigate the effect of stromal-derived factor-1-alpha (SDF-1-alpha) on endothelial angiogenic effects. SDF-1-alpha (50 ng/ml) increased the number of cultured endothelial cells from 33,653 +/- 1183 to 55,398 +/- 2741, which significantly reduced by adding the BK(Ca)-inhibitor iberiotoxin, or the endothelial nitric oxide synthase-blocker, L-NMMA (n = 24, p < 0.05). Using the "Fences"-assay a significant increase of HUVEC migration induced by SDF-1-alpha was reported, which was blocked by the addition of iberiotoxin or L-NMMA (n = 12, p < 0.05). BK(Ca) open-state probability (NPo) was analysed using the patch-clamp technique and NPo was increased from 0.003 (control) to 0.052 (SDF-1-alpha; n = 10, p < 0.05). NO synthesis was measured using a cGMP-radioimmunoassay. A significant increase of cGMP levels from 0.952 pmol/mg protein to 2.179 pmol/mg protein was observed, that was abolished by L-NMMA and significantly reduced by iberiotoxin (n=15, p<0.05). SDF-1-alpha increases endothelial proliferation and migration involving the activation of BK(Ca) and an increased production of NO.
- Published
- 2005
- Full Text
- View/download PDF
30. Margatoxin inhibits VEGF-induced hyperpolarization, proliferation and nitric oxide production of human endothelial cells.
- Author
-
Erdogan A, Schaefer CA, Schaefer M, Luedders DW, Stockhausen F, Abdallah Y, Schaefer C, Most AK, Tillmanns H, Piper HM, and Kuhlmann CR
- Subjects
- Calcium metabolism, Cell Division drug effects, Cell Polarity drug effects, Cell Survival drug effects, Cells, Cultured, Cyclic GMP metabolism, Dose-Response Relationship, Drug, Drug Interactions, Endothelium, Vascular metabolism, Enzyme Inhibitors pharmacology, Humans, Kv1.3 Potassium Channel, Potassium Channels, Voltage-Gated metabolism, Scorpion Venoms, Umbilical Veins cytology, Vascular Endothelial Growth Factor Receptor-2 metabolism, omega-N-Methylarginine pharmacology, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Neurotoxins pharmacology, Nitric Oxide metabolism, Vascular Endothelial Growth Factor A pharmacology
- Abstract
Background: Vascular endothelial growth factor (VEGF) induces proliferation of endothelial cells (EC) in vitro and angiogenesis in vivo. Furthermore, a role of VEGF in K(+) channel, nitric oxide (NO) and Ca(2+) signaling was reported. We examined whether the K(+) channel blocker margatoxin (MTX) influences VEGF-induced signaling in human EC., Methods: Fluorescence imaging was used to analyze changes in the membrane potential (DiBAC), intracellular Ca(2+) (FURA-2) and NO (DAF) levels in cultured human EC derived from human umbilical vein EC (HUVEC). Proliferation of HUVEC was examined by cell counts (CC) and [(3)H]-thymidine incorporation (TI)., Results: VEGF (5--50 ng/ml) caused a dose-dependent hyperpolarization of EC, with a maximum at 30 ng/ml (n=30, p<0.05). This effect was completely blocked by MTX (5 micromol/l). VEGF caused an increase in transmembrane Ca(2+) influx (n=30, p<0.05) that was sensitive to MTX and the blocker of transmembrane Ca(2+) entry 2-aminoethoxydiphenyl borate (APB, 100 micromol/l). VEGF-induced NO production was significantly reduced by MTX, APB and a reduction in extracellular Ca(2+) (n=30, p<0.05). HUVEC proliferation, examined by CC and TI, was significantly increased by VEGF and inhibited by MTX (CC: -58%, TI --121%); APB (CC --99%, TI--187%); N-monomethyl-L-arginine (300 micromol/l: CC: -86%, TI --164%)., Conclusions: VEGF caused an MTX-sensitive hyperpolarization which results in an increased transmembrane Ca(2+) entry that is responsible for the effects on endothelial proliferation and NO production., (Copyright (c) 2005 S. Karger AG, Basel.)
- Published
- 2005
- Full Text
- View/download PDF
31. A new signaling mechanism of hepatocyte growth factor-induced endothelial proliferation.
- Author
-
Kuhlmann CR, Schaefer CA, Fehsecke A, Most AK, Tillmanns H, and Erdogan A
- Subjects
- Dose-Response Relationship, Drug, Humans, Membrane Potentials, Mitogen-Activated Protein Kinases metabolism, Nitric Oxide biosynthesis, Phosphorylation, Potassium Channels physiology, Umbilical Veins cytology, Cell Proliferation, Endothelium, Vascular cytology, Hepatocyte Growth Factor pharmacology, Signal Transduction
- Abstract
Background: The hepatocyte growth factor (HGF) has been shown to promote endothelial cell proliferation. In this study, the signaling cascade responsible for the HGF-induced proliferation was examined., Methods: The proliferation of human umbilical cord vein endothelial cells (HUCVEC) was determined using cell counts. Changes of the membrane potential were analyzed using the fluorescence dye DiBAC. Intracellular cGMP-levels were measured by means of [3H]-cGMP-radioimmunoassay. Phosphorylation of the p42/p44 MAP-kinase (MAPK) and the endothelial nitric oxide synthase (eNOS) was analyzed by immunocytochemistry., Results: A dose-dependent (1-30 ng mL(-1)) increase of HUCVEC proliferation with a maximum at a concentration of 15 ng mL(-1) was induced by HGF. This effect was significantly reduced by the addition of the K+ channel blocker iberiotoxin (100 nmol L(-1)), eNOS inhibitor L-NMMA (300 micromol L(-1)), or the MEK inhibitor PD 98059 (20 micromol L(-1)). A HGF-induced hyperpolarization that was blocked by iberiotoxin was observed. In addition, HGF-induced activation of the eNOS was blocked by the K+ channel inhibitor. An increase of +101% MAPK phosphorylation was induced by HGF, which was blocked, if the cells were treated with L-NMMA (n = 20; P < 0.05), whereas HGF-induced phosphorylation of the eNOS was not affected by MEK inhibition., Conclusions: Hepatocyte growth factor modulates endothelial K+ channels causing an activation of the eNOS; the increase of nitric oxide is necessary for the phosphorylation of the MAPK inducing the proliferation of HUCVEC.
- Published
- 2005
- Full Text
- View/download PDF
32. Quercetin-induced induction of the NO/cGMP pathway depends on Ca2+-activated K+ channel-induced hyperpolarization-mediated Ca2+-entry into cultured human endothelial cells.
- Author
-
Kuhlmann CR, Schaefer CA, Kosok C, Abdallah Y, Walther S, Lüdders DW, Neumann T, Tillmanns H, Schäfer C, Piper HM, and Erdogan A
- Subjects
- Cell Division drug effects, Cyclic GMP biosynthesis, Dose-Response Relationship, Drug, Endothelial Cells drug effects, Endothelial Cells metabolism, Endothelial Cells physiology, Humans, Membrane Potentials, Nitric Oxide biosynthesis, Potassium Channels, Calcium-Activated physiology, Quercetin administration & dosage, Quercetin therapeutic use, Calcium metabolism, Phytotherapy, Plants, Medicinal, Potassium Channels, Calcium-Activated drug effects, Quercetin pharmacology
- Abstract
Quercetin is one of the dietary-derived flavonoids that are held responsible for the beneficial effects of red wine drinking in coronary artery disease known as the "French paradox". We examined whether quercetin modulates endothelial function by influencing Ca2+-activated K+ channels with large conductance (BK(Ca)) in cultured human endothelial cells. Membrane potential and intracellular Ca2+ concentrations of cultured human endothelial cells derived from umbilical cord veins (HUVEC) were measured using the fluorescence dyes DiBAC, and FURA-2, respectively. NO production was examined using a cGMP radioimmunoassay. HUVEC proliferation was analyzed by cell counts and thymidine incorporation. A dose-dependent hyperpolarization of HUVEC was recorded when quercetin was added (5-100 micromol/L). The maximum effect (50 micromol/L) was significantly reduced by the addition of the highly selective BK(Ca) inhibitor iberiotoxin (100 nmol/L), but not by blockers of other Ca2+-activated K+ channels (n = 30; p < 0.05). This BK(Ca)-induced hyperpolarization caused a transmembrane Ca2+ influx, because the quercetin-induced increase of intracellular Ca2+ was blocked by iberiotoxin, or by applying 2-aminoethoxydiphenylborate (100 micromol/L)--an inhibitor of capacitative Ca2+ entry (n = 30; p < 0.05). Quercetin-induced cGMP levels were significantly reduced by the eNOS-inhibitor l-NMMA (300 micromol/L), and by iberiotoxin (n = 10; p < 0.05). Endothelial proliferation was significantly reduced by 56 % when cells were incubated with quercetin (n = 12; p < 0.05). This effect was due to the increased NO production, because it was reversed when the cells were treated with a combination of quercetin and l-NMMA. In conclusion quercetin improves endothelial dysfunction by increasing NO synthesis involving BK(Ca)-dependent membrane hyperpolarization-induced capacitative Ca 2+ entry. Increased NO production is responsible for the quercetin-dependent inhibition of endothelial proliferation.
- Published
- 2005
- Full Text
- View/download PDF
33. Discordant effects of nicotine on endothelial cell proliferation, migration, and the inward rectifier potassium current.
- Author
-
Kuhlmann CR, Scharbrodt W, Schaefer CA, Most AK, Backenköhler U, Neumann T, Tillmanns H, Waldecker B, Erdogan A, and Wiecha J
- Subjects
- Cell Proliferation drug effects, Cells, Cultured, Cyclic GMP metabolism, Electric Conductivity, Endothelial Cells metabolism, Fibroblast Growth Factor 2 antagonists & inhibitors, Fibroblast Growth Factor 2 pharmacology, Humans, Ion Transport drug effects, Umbilical Veins cytology, Cell Movement drug effects, Endothelial Cells cytology, Endothelial Cells drug effects, Nicotine pharmacology, Potassium metabolism
- Abstract
The inward rectifier K+ current (K(ir)) determines the resting membrane potential of endothelial cells. Basic fibroblast growth factor (bFGF) has been shown to activate K(ir) and acts as angiogenic factor and vasodilator. In contrast, nicotine has been demonstrated to reduce endothelium-dependent vasorelaxation by increasing radical formation. Aim of the present study was to investigate whether nicotine modulates K(ir) and if this plays a role in bFGF-mediated proliferation, migration and nitric oxide (NO)-formation of endothelial cells. Using the patch-clamp technique in cultured endothelial cells of human umbilical cord veins (HUVEC), we found characteristic K(ir), which were blocked by extracellular barium (100 micromol/l). Perfusion with nicotine (1 nmol/l-10 micromol/l) revealed a dose-dependent reduction of K(ir). The simultaneous perfusion with bFGF (50 ng/ml) and nicotine (10 micromol/l) still significantly reduced K(ir) (n = 8; P < 0.01). Cell counts revealed that bFGF-mediated proliferation of HUVEC was significantly inhibited when using 1-10 micromol/l nicotine (n = 8, P < 0.01). The bFGF-induced endothelial cell migration--examined using the "Fences-Migration-Assay"--was significantly reduced by 10 mumol/l nicotine (n = 12; P < 0.05). NO-production was examined using a cGMP-Radioimmunoassay. The significant bFGF-induced increase of cGMP-levels was reduced by nicotine (n = 10; P < 0.05). Our data indicate that the modulation of K(ir) seems to be an essential pathway in the antagonistic effects of nicotine on bFGF-mediated endothelial cell growth, migration and NO-formation.
- Published
- 2005
- Full Text
- View/download PDF
34. Endothelin-1-induced proliferation of human endothelial cells depends on activation of K+ channels and Ca+ influx.
- Author
-
Kuhlmann CR, Most AK, Li F, Münz BM, Schaefer CA, Walther S, Raedle-Hurst T, Waldecker B, Piper HM, Tillmanns H, and Wiecha J
- Subjects
- Calcium metabolism, Calcium physiology, Cell Count, Cell Division drug effects, Cell Division physiology, Cells, Cultured, Chelating Agents pharmacology, Culture Media, Dose-Response Relationship, Drug, Egtazic Acid pharmacology, Electric Conductivity, Endothelial Cells drug effects, Endothelin B Receptor Antagonists, Humans, Membrane Potentials physiology, Oligopeptides, Peptides pharmacology, Piperidines, Potassium Channels, Calcium-Activated antagonists & inhibitors, Egtazic Acid analogs & derivatives, Endothelial Cells physiology, Endothelin-1 physiology, Potassium Channels, Calcium-Activated physiology
- Abstract
Aims: Endothelin-1 (ET-1) promotes endothelial cell growth. Endothelial cell proliferation involves the activation of Ca2+-activated K+ channels. In this study, we investigated whether Ca2+-activated K+ channels with big conductance (BK(Ca)) contribute to endothelial cell proliferation induced by ET-1., Methods: The patch-clamp technique was used to analyse BK(Ca) activity in endothelial cells derived from human umbilical cord veins (HUVEC). Endothelial proliferation was examined using cell counts and measuring [3H]-thymidine incorporation. Changes of intracellular Ca2+ levels were examined using fura-2 fluorescence imaging., Results: Characteristic BK(Ca) were identified in cultured HUVEC. Continuous perfusion of HUVEC with 10 nmol L(-1) ET-1 caused a significant increase of BK(Ca) open-state probability (n = 14; P < 0.05; cell-attached patches). The ET(B)-receptor antagonist (BQ-788, 1 micromol L(-1)) blocked this effect. Stimulation with Et-1 (10 nmol L(-1)) significantly increased cell growth by 69% (n = 12; P < 0.05). In contrast, the combination of ET-1 (10 nmol L(-1)) and the highly specific BK(Ca) blocker iberiotoxin (IBX; 100 nmol L(-1)) did not cause a significant increase in endothelial cell growth. Ca2+ dependency of ET-1-induced proliferation was tested using the intracellular Ca2+-chelator BAPTA (10 micromol L(-1)). BAPTA abolished ET-1 induced proliferation (n = 12; P < 0.01). In addition, ET-1-induced HUVEC growth was significantly reduced, if cells were kept in a Ca2+-reduced solution (0.3 mmol L(-1)), or by the application of 2 aminoethoxdiphenyl borate (100 micromol L(-1)) which blocks hyperpolarization-induced Ca2+ entry (n = 12; P < 0.05)., Conclusion: Activation of BK(Ca) by ET-1 requires ET(B)-receptor activation and induces a capacitative Ca2+ influx which plays an important role in ET-1-mediated endothelial cell proliferation.
- Published
- 2005
- Full Text
- View/download PDF
35. The K+-channel opener NS1619 increases endothelial NO-synthesis involving p42/p44 MAP-kinase.
- Author
-
Kuhlmann CR, Trümper JR, Abdallah Y, Wiebke Lüdders D, Schaefer CA, Most AK, Backenköhler U, Neumann T, Walther S, Piper HM, Tillmanns H, and Erdogan A
- Subjects
- Calcium metabolism, Cell Proliferation, Cells, Cultured, Humans, Large-Conductance Calcium-Activated Potassium Channels, Nitric Oxide Synthase metabolism, Nitric Oxide Synthase Type III, Phosphorylation, Potassium Channels, Calcium-Activated drug effects, Umbilical Veins, Benzimidazoles pharmacology, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, Nitric Oxide biosynthesis, Potassium Channels, Calcium-Activated physiology
- Abstract
Ca(2+)-activated K(+) channels with large conductance (BK(Ca)) have been shown to play an important role in the regulation of vascular tone. We examined the role of the p42/p44 MAP-kinase (p42/p44(MAPK)) on nitric oxide (NO) production in human endothelial cells induced by the BK(Ca)-opener NS1619. Using DiBAC-fluorescence imaging a concentration-dependent (2.5-12.5 microM) hyperpolarization induced by NS1619 was observed. A significant increase of intracellular Ca(2+)-concentration by NS1619 was seen using Fura-2-fluorescence-imaging, which was blocked by 2-APB, or reduction of extracellular Ca(2+) (n=30; p<0.05). A cGMP-radioimmunoassay was used to examine NO synthesis. NS1619 significantly increased cGMP levels which was inhibited by LNMMA, iberiotoxin, BAPTA, 2-APB, reduction of extracellular Ca(2+), PD 98059, or U0126 (cGMP (pmol/mg protein): NS1619 3.25 +/- 0.85; NS1619 + L-NMMA 0.86 +/- 0.02; NS1619 + iberiotoxin 0.99 +/- 0.09; NS1619 + BAPTA 0.93 +/- 0.29; NS1619 + 2-APB 0.99 +/- 0.31; NS1619 + Ca(2+)-reduction 1.17 +/- 0.06; NS1619 + PD98059 1.06 +/- 0.49; NS1619 + U0126 1.10 +/- 0.24; n=10; p<0.05). The phosphorylation of eNOS and p42/p44(MAPK) was examined by immunocytochemistry. Phosphorylation of p42/p44(MAPK) was significantly increased after 10 minutes of NS1619 stimulation, whereas eNOS phosphorylation was not changed over a period of 1 to 30 minutes. NS1619-induced hyperpolarization was not affected by treatment with PD 98059 or U0126. Additionally, NS1619 inhibited endothelial proliferation involving a NO-dependent mechanism. Our data demonstrate that NS1619 causes a transmembrane Ca(2+)-influx leading to an increased NO production involving p42/p44(MAPK). This rise of NO formation is responsible for the NS1619 induced reduction of endothelial cell growth.
- Published
- 2004
- Full Text
- View/download PDF
36. bFGF activates endothelial Ca2+-activated K+ channels involving G-proteins and tyrosine kinases.
- Author
-
Kuhlmann CR, Wu Y, Li F, Münz BM, Tillmanns H, Waldecker B, and Wiecha J
- Subjects
- Cell Proliferation, Cells, Cultured, Coronary Vessels cytology, Coronary Vessels enzymology, Female, Humans, Membrane Potentials physiology, Middle Aged, Patch-Clamp Techniques, Signal Transduction physiology, Endothelial Cells enzymology, Fibroblast Growth Factor 2 metabolism, GTP-Binding Proteins metabolism, Potassium Channels, Calcium-Activated metabolism, Protein-Tyrosine Kinases metabolism
- Abstract
Activation of Ca2+-activated K+ channels (BK(Ca)) has been shown to be an important step in the basic fibroblast growth factor (bFGF)-induced proliferation of endothelial cells. In this study, we investigate the signaling cascades of BK(Ca) modulation by bFGF. Using the patch-clamp technique, bFGF (50 ng/ml) significantly increased the BK(Ca) open-state probability in cultured endothelial cells derived from human coronary arteries after 6 min (n=26, p<0.01), which lasted up the whole recording time of 60 min. After preincubation with pertussis toxin (100 ng/ml), bFGF superfusion did not cause a significant increase of BK(Ca) activity until 25 min had passed. When genistein was supplemented to the bath solution, a significant activation of BK(Ca) by bFGF was observed during a time interval of 6-20 min (n=17, p<0.01). In contrast, the addition of the inactive analogue daidzein did not change bFGF-induced activation of the BK(Ca). In conclusion, the results of the present study indicate that the early activation of the BK(Ca) by bFGF is mediated by G-protein-dependent mechanisms, whereas the later effect is due to a tyrosine kinase-dependent signaling pathway.
- Published
- 2004
- Full Text
- View/download PDF
37. Dose-dependent activation of Ca2+-activated K+ channels by ethanol contributes to improved endothelial cell functions.
- Author
-
Kuhlmann CR, Li F, Lüdders DW, Schaefer CA, Most AK, Backenköhler U, Neumann T, Tillmanns H, Waldecker B, Erdogan A, and Wiecha J
- Subjects
- Cell Proliferation drug effects, Cells, Cultured, Dose-Response Relationship, Drug, Humans, Nitric Oxide physiology, Endothelium, Vascular drug effects, Endothelium, Vascular physiology, Ethanol pharmacology, Potassium Channels, Calcium-Activated physiology
- Abstract
Background: Regular moderate alcohol (EtOH) intake seems to protect against both coronary artery disease and ischemic stroke, whereas the risk increases with heavy EtOH consumption. Effects of EtOH on endothelial cell function may be relevant to these disparate effects. Potassium channels play an important role in the regulation of endothelial cell functions. Therefore, we investigated whether Ca-activated K channels (BKCa) are modulated by EtOH. Furthermore, we examined whether EtOH-induced changes of endothelial nitric oxide (NO) formation and cell proliferation are due to BKCa activation., Methods: The patch-clamp technique was used to investigate BKCa activity in cultured human umbilical vein endothelial cells (HUVEC). NO formation was analyzed by using the fluorescence dye 4,5-diaminofluorescein. Endothelial proliferation was examined by using cell counts and measuring [H]thymidine incorporation., Results: EtOH dose-dependently (10-150 mmol/liter) modulated BKCa-activity, with the highest increase of open-state probability at a concentration of 50 mmol/liter (n = 13; p < 0.05). Inside-out recordings revealed that this effect was due to direct BKCa activation, whereas open-state probability was not changed in cell-attached recordings after pertussis toxin preincubation. EtOH (10 and 50 mmol/liter) caused a significant increase of NO levels, which was blocked by the highly selective BKCa inhibitor iberiotoxin (100 nmol/l; n = 30; p < 0.05). Higher concentrations of EtOH (100 and 150 mmol/liter) significantly reduced NO synthesis (n = 30; p < 0.05). Both methods revealed a significant increase of HUVEC proliferation, which was inhibited by iberiotoxin (n = 30; p < 0.05). At a concentration of 150 mmol/liter, EtOH caused a significant reduction of endothelial proliferation., Conclusions: EtOH directly activates BKCa in HUVEC, leading to an increase of endothelial proliferation and production of NO. These results indicate a possible beneficial effect of low-dose EtOH on endothelial function, whereas higher concentrations must be considered as harmful.
- Published
- 2004
- Full Text
- View/download PDF
38. Basic fibroblast growth factor-induced endothelial proliferation and NO synthesis involves inward rectifier K+ current.
- Author
-
Scharbrodt W, Kuhlmann CR, Wu Y, Schaefer CA, Most AK, Backenköhler U, Neumann T, Tillmanns H, Waldecker B, Erdogan A, and Wiecha J
- Subjects
- Barium metabolism, Barium pharmacology, Calcium metabolism, Cell Division drug effects, Cells, Cultured cytology, Cells, Cultured drug effects, Cells, Cultured physiology, Cyclic GMP biosynthesis, Endothelial Cells cytology, Endothelial Cells physiology, Humans, Ion Channel Gating drug effects, Ion Channel Gating physiology, Ion Transport drug effects, Ion Transport physiology, Membrane Potentials drug effects, Membrane Potentials physiology, Patch-Clamp Techniques, Potassium metabolism, Endothelial Cells drug effects, Fibroblast Growth Factor 2 pharmacology, Nitric Oxide biosynthesis, Potassium Channels, Inwardly Rectifying physiology
- Abstract
Objective: Inward rectifier K+ currents (K(ir)) determine the resting membrane potential and thereby modulate essential Ca2+-dependent pathways, like cell growth and synthesis of vasoactive agents in endothelial cells. Basic fibroblast growth factor (bFGF) acts as a vasodilatator and angiogenic factor. Therefore, we investigated the effect of bFGF on K(ir) and assessed the role in proliferation and nitric oxide (NO) formation of endothelial cells., Methods and Results: Using the patch-clamp technique, we found characteristic K(ir) in human umbilical cord vein endothelial cells (HUVEC), which were dose-dependently blocked by barium (10 to 100 micromol/L). Perfusion with bFGF (50 ng/mL) caused a significant increase of K(ir), which was blocked by 100 micromol/L barium (n=18, P<0.01). The bFGF-induced HUVEC proliferation was significantly inhibited when using 50 to 100 micromol/L barium (n=6; P<0.01). NO production was examined using a cGMP radioimmunoassay. bFGF caused a significant increase of cGMP levels (n=10; P<0.05), which were blocked by barium., Conclusions: Modulation of K(ir) plays an important role in bFGF-mediated endothelial cell growth and NO formation.
- Published
- 2004
- Full Text
- View/download PDF
39. Lysophosphatidylcholine-induced modulation of Ca(2+)-activated K(+)channels contributes to ROS-dependent proliferation of cultured human endothelial cells.
- Author
-
Wolfram Kuhlmann CR, Wiebke Lüdders D, Schaefer CA, Kerstin Most A, Backenköhler U, Neumann T, Tillmanns H, and Erdogan A
- Subjects
- Calcium metabolism, Cell Proliferation drug effects, Cells, Cultured, Cyclic GMP metabolism, Endothelial Cells cytology, Endothelial Cells metabolism, Homeostasis drug effects, Humans, NADPH Oxidases metabolism, Nitric Oxide Synthase metabolism, Nitric Oxide Synthase Type II, Nitrogen Oxides metabolism, Umbilical Cord drug effects, Endothelial Cells drug effects, Lysophosphatidylcholines pharmacology, Potassium Channels, Calcium-Activated metabolism, Reactive Oxygen Species metabolism, Umbilical Cord cytology, Umbilical Cord metabolism
- Abstract
Proliferation of endothelial cells plays a crucial role in the process of atherosclerotic plaque destabilization. The major component of oxidized low-density lipoprotein lysophosphatidylcholine (LPC) has been shown to promote endothelial proliferation by increasing the production of reactive oxygen species (ROS). Since K(+) channels are known to control the cell cycle, we investigated the role of Ca(2+)-activated K(+) channels (BK(Ca)) in the regulation of LPC-induced endothelial proliferation and ROS generation. A significant increase of cell growth induced by LPC (20 micromol/l; cell counts (CCs): +87%, thymidin incorporation: +89%; n = 12, P < 0.01) was observed, which was inhibited by the BK(Ca) inhibitor iberiotoxin (IBX; 100 nmol/l), by the NAD(P)H-oxidase inhibitor diphenyleneiodonium (5 micromol/l) and by transfection with antisense (AS) oligonucleotides against NAD(P)H oxidase, whereas N(G)-monomethyl-l-arginine (l-NMMA) further increased LPC-induced cell growth. Using the patch-clamp technique a significant increase of BK(Ca) open-state probability (control: 0.004 +/- 0.002; LPC: 0.104 +/- 0.035; n = 21, P < 0.05) by LPC was observed. Using dichlorofluorescein fluorescence microscopy a significant increase of ROS induced by LPC was reported, that was blocked by IBX and Ca(2+) antagonists. Intracellular Ca(2+) measurements revealed a capacitative Ca(2+) influx caused by LPC. Bioactivity of nitric oxide (NO) was measured using a [(3)H]-cGMP radioimmunoassay. LPC significantly decreased acetylcholine-induced NO synthesis. LPC significantly increased cGMP levels in endothelial cells transfected with AS, which was blocked by IBX. In conclusion, our results demonstrate that LPC activates BK(Ca) thereby increasing ROS production which induces endothelial proliferation. In addition LPC-induced BK(Ca)-activation contributes to increased cGMP levels, if ROS production is prevented by AS.
- Published
- 2004
- Full Text
- View/download PDF
40. Cerivastatin activates endothelial calcium-activated potassium channels and thereby modulates endothelial nitric oxide production and cell proliferation.
- Author
-
Kuhlmann CR, Gast C, Li F, Schäfer M, Tillmanns H, Waldecker B, and Wiecha J
- Subjects
- Cell Division drug effects, Cells, Cultured, Humans, Endothelium drug effects, Endothelium metabolism, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Nitric Oxide biosynthesis, Potassium Channels, Calcium-Activated drug effects, Pyridines pharmacology
- Abstract
Statins are known to counteract the process of arteriosclerosis by exerting direct pleiotropic effects on vascular endothelium. The aim of this study was to investigate a possible effect of cerivastatin on endothelial Ca(2+)-activated K+ channels (BK(Ca)) and to assess their contribution to cerivastatin-mediated changes of endothelial nitric oxide (NO) production and proliferation. Membrane potential was measured using bis-1,3-dibutylbarbituric acid-trimethine oxonol-fluorescence imaging. Patch-clamp recordings of BK(Ca) were performed on cultured human umbilical vein endothelial cells. NO production was measured using 4,5-diaminofluorescein-fluorescence imaging and a [(3)H]cGMP RIA. Proliferation was analyzed by means of cell counts and [(3)H]thymidine incorporation (TI). Cerivastatin (0.001 to 0.05 micromol/L) caused a significant membrane hyperpolarization (n = 30; P < 0.05). This effect was abolished using the BK(Ca) inhibitor iberiotoxin (IBX; 100 nmol/L). The addition of mevalonate (500 micromol/L) blocked the BK(Ca) activation induced by cerivastatin (n = 19; P < 0.05). Endothelial cGMP level was increased by acetylcholine (ACh; 1 micromol/L). The combination of ACh and cerivastatin additionally increased cGMP levels, with a maximum at 0.03 micromol/L cerivastatin (84%; n = 10, P < 0.01). ACh-induced increase of cGMP-level was significantly reduced by IBX (n = 10, P < 0.01) as it was with all combined administrations of ACh and cerivastatin. 4,5-Diaminofluorescein-fluorescence measurements revealed a significant increase of NO levels by cerivastatin, which was abolished by IBX (n = 30; P < 0.05). Cell counts and TI demonstrated significant inhibition of human umbilical vein endothelial cell proliferation with a maximum at 0.03 micro mol/L (cell count, -32.2%; TI, -70%; n = 12; P < 0.01). These data show that cerivastatin activates endothelial BK(Ca), which plays an important role in the signaling of cerivastatin-mediated endothelial NO production and proliferation.
- Published
- 2004
- Full Text
- View/download PDF
41. Statins prevent oxidized low-density lipoprotein- and lysophosphatidylcholine-induced proliferation of human endothelial cells.
- Author
-
Schaefer CA, Kuhlmann CR, Gast C, Weiterer S, Li F, Most AK, Neumann T, Backenköhler U, Tillmanns H, Waldecker B, Wiecha J, and Erdogan A
- Subjects
- Cell Division drug effects, Cell Division physiology, Cells, Cultured, Dose-Response Relationship, Drug, Endothelium, Vascular cytology, Humans, Lipoproteins, LDL antagonists & inhibitors, Lysophosphatidylcholines antagonists & inhibitors, Endothelium, Vascular drug effects, Growth Inhibitors pharmacology, Hydroxymethylglutaryl-CoA Reductase Inhibitors pharmacology, Lipoproteins, LDL pharmacology, Lysophosphatidylcholines pharmacology
- Abstract
The proliferation of endothelial cells is induced by oxidized low-density lipoprotein (oxLDL) and its major component, lysophosphatidylcholine (LPC). The aim of this study was to investigate the effect of statins on the proliferation of endothelial cells derived from human umbilical cord veins (HUVEC). Cerivastatin, simvastatin and fluvastatin caused a dose-dependent inhibition of endothelial cell growth (n=12; P<.01) when using cell counts and [3H]-thymidine incorporation, respectively. The strongest inhibition of HUVEC proliferation was achieved at statin concentrations of 0.1 micromol/l (cerivastatin), 2.5 micromol/l (simvastatin) and 1 micromol/l (fluvastatin). Cell counts were significantly reduced from 22937+/-280.6 (control) to 7791+/-133.6 (cerivastatin), 7292+/-146.6 (simvastatin) and 6792+/-135.5 (fluvastatin) (n=12; P<.01). Interestingly, cell proliferation induced by oxLDL (10 microg/ml) and LPC (20 micromol/l) could be effectively prevented using statins at concentrations between 0.01 and 0.1 micromol/l (cerivastatin), 1 and 2.5 micromol/l (simvastatin) and 0.25 and 1 micromol/l (fluvastatin). This effect of the statins was abolished by preincubation with mevalonate (500 micromol/l). Our results demonstrate an interesting direct effect of statins on the proliferation of human endothelial cells induced by oxLDL and LPC, which may be beneficial to prevent vascular effects of these atherogenic lipids.
- Published
- 2004
- Full Text
- View/download PDF
42. Modulation of endothelial Ca(2+)-activated K(+) channels by oxidized LDL and its contribution to endothelial proliferation.
- Author
-
Kuhlmann CR, Schäfer M, Li F, Sawamura T, Tillmanns H, Waldecker B, and Wiecha J
- Subjects
- Acetylcholine pharmacology, Arteriosclerosis pathology, Calcium metabolism, Cell Division drug effects, Cells, Cultured, Endothelium, Vascular pathology, Humans, Lipoproteins, LDL metabolism, Nitric Oxide metabolism, Patch-Clamp Techniques, Arteriosclerosis metabolism, Endothelium, Vascular metabolism, Lipoproteins, LDL pharmacology, Potassium Channels, Calcium-Activated metabolism
- Abstract
Objective: Oxidized low-density lipoprotein (oxLDL) plays an important role in causing endothelial dysfunction and initiating atherosclerosis. Some of the endothelial functions have been shown to be modulated by changes in cellular electrophysiological properties. Therefore, we analysed the effect of oxLDL on endothelial Ca(2+)-activated K(+) channels (BK(Ca)) and its contribution to oxLDL-mediated changes of proliferation and syntheses of nitric oxide (NO)., Methods: The patch-clamp technique was used to study the behavior of BK(Ca) in human endothelial cells of umbilical cord veins (HUVEC). Changes of intracellular Ca(2+) were measured by means of Fura-2 imaging. Cell counts and [3H]-thymidine incorporation were used to analyse endothelial proliferation. Synthesis of NO was measured by means of [3H]-cGMP radioimmunoassay., Results: oxLDL (10 microg/ml) caused a significant increase of BK(Ca) activity, whereas preincubation of HUVEC with an antibody against the lectin-like-oxLDL-receptor-1 (LOX-1) abolished BK(Ca) activation. Fura-2 measurements revealed a biphasic increase of intracellular Ca(2+) after application of the atherogenic lipid. Endothelial proliferation was significantly increased by oxLDL. The highly selective BK(Ca) inhibitor iberiotoxin (100 nmol/l IBX) blocked this proliferative response. Acetylcholine-induced NO synthesis was significantly decreased by IBX. Interestingly, oxLDL significantly decreased acetylcholine-induced NO synthesis if the production of superoxide was not blocked by antisense oligonucleotides against the NAD(P)H-oxidase., Conclusions: Our data demonstrate that oxLDL activates BK(Ca), which plays an important role in oxLDL-mediated endothelial proliferation. Acetylcholine-induced NO synthesis is modulated by BK(Ca), whereas the reduction of acetylcholine-induced NO-synthesis by oxLDL is related to an increase in superoxide production.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.