4 results on '"Loop, Torsten"'
Search Results
2. Carbon monoxide abrogates ischemic insult to neuronal cells via the soluble guanylate cyclase-cGMP pathway
- Author
-
Schallner, Nils, Romão, Carlos C., Biermann, Julia, Lagrèze, Wolf A., Otterbein, Leo E., Buerkle, Hartmut, Loop, Torsten, and Goebel, Ulrich
- Subjects
Male ,Retinal Ganglion Cells ,Receptors, Cytoplasmic and Nuclear ,lcsh:Medicine ,Apoptosis ,Signal transduction ,Brain Ischemia ,Rats, Sprague-Dawley ,Molecular cell biology ,Soluble Guanylyl Cyclase ,Coordination Complexes ,lcsh:Science ,Cyclic GMP ,Cellular Stress Responses ,Neurons ,Carbon Monoxide ,Cell Death ,Neurochemistry ,Animal Models ,Flow Cytometry ,Neuroprotective Agents ,Neurology ,Reperfusion Injury ,Medicine ,Retinal Disorders ,Female ,Neurochemicals ,Research Article ,Drugs and Devices ,Cerebrovascular Diseases ,Signaling in cellular processes ,Nitric Oxide ,Signaling Pathways ,Gene Expression Regulation, Enzymologic ,Model Organisms ,Neuropharmacology ,Cell Line, Tumor ,Rotenone ,Organometallic Compounds ,Animals ,Humans ,Biology ,Ischemic Stroke ,Molybdenum ,Dose-Response Relationship, Drug ,lcsh:R ,NADPH Oxidases ,Rats ,Ophthalmology ,Guanylate Cyclase ,cGMP signaling ,Neuro-Ophthalmology ,Rat ,lcsh:Q ,Antiapoptotic signaling ,Molecular Neuroscience ,Reactive Oxygen Species ,Cytometry ,Neuroscience - Abstract
Purpose Carbon monoxide (CO) is an accepted cytoprotective molecule. The extent and mechanisms of protection in neuronal systems have not been well studied. We hypothesized that delivery of CO via a novel releasing molecule (CORM) would impart neuroprotection in vivo against ischemia-reperfusion injury (IRI)-induced apoptosis of retinal ganglion cells (RGC) and in vitro of neuronal SH-SY5Y-cells via activation of soluble guanylate-cyclase (sGC). Methods To mimic ischemic respiratory arrest, SH-SY5Y-cells were incubated with rotenone (100 nmol/L, 4 h) ± CORM ALF186 (10–100 µmol/L) or inactivated ALF186 lacking the potential of releasing CO. Apoptosis and reactive oxygen species (ROS) production were analyzed using flow-cytometry (Annexin V, mitochondrial membrane potential, CM-H2DCFDA) and Western blot (Caspase-3). The impact of ALF186± respiratory arrest on cell signaling was assessed by measuring expression of nitric oxide synthase (NOS) and soluble guanylate-cyclase (sGC) and by analyzing cellular cGMP levels. The effect of ALF186 (10 mg/kg iv) on retinal IRI in Sprague-Dawley rats was assessed by measuring densities of fluorogold-labeled RGC after IRI and by analysis of apoptosis-related genes in retinal tissue. Results ALF186 but not inactivated ALF186 inhibited rotenone-induced apoptosis (Annexin V positive cells: 25±2% rotenone vs. 14±1% ALF186+rotenone, p
- Published
- 2013
3. Postconditioning with Inhaled Carbon Monoxide Counteracts Apoptosis and Neuroinflammation in the Ischemic Rat Retina.
- Author
-
Schallner, Nils, Fuchs, Matthias, Schwer, Christian I., Loop, Torsten, Buerkle, Hartmut, Lagrèze, Wolf Alexander, van Oterendorp, Christian, Biermann, Julia, Goebel, Ulrich, and Barnes, Steven
- Subjects
ISCHEMIA ,REPERFUSION injury ,CELL death ,MICROGLIA ,REPERFUSION ,ORAL mucosa - Abstract
Purpose: Ischemia and reperfusion injury (I/R) of neuronal structures and organs is associated with increased morbidity and mortality due to neuronal cell death. We hypothesized that inhalation of carbon monoxide (CO) after I/R injury ('postconditioning') would protect retinal ganglion cells (RGC). Methods: Retinal I/R injury was performed in Sprague-Dawley rats (n = 8) by increasing ocular pressure (120 mmHg, 1 h). Rats inhaled room air or CO (250 ppm) for 1 h immediately following ischemia or with 1.5 and 3 h latency. Retinal tissue was harvested to analyze Bcl-2, Bax, Caspase-3, HO-1 expression and phosphorylation of the nuclear transcription factor (NF)-κB, p38 and ERK-1/2 MAPK. NF-κB activation was determined and inhibition of ERK-1/2 was performed using PD98059 (2 mg/ kg). Densities of fluorogold prelabeled RGC were analyzed 7 days after injury. Microglia, macrophage and Müller cell activation and proliferation were evaluated by Iba-1, GFAP and Ki-67 staining. Results: Inhalation of CO after I/R inhibited Bax and Caspase-3 expression (Bax: 1.9±0.3 vs. 1.4±0.2, p = 0.028; caspase-3: 2.0±0.2 vs. 1.5±0.1, p = 0.007; mean±S.D., fold induction at 12 h), while expression of Bcl-2 was induced (1.2±0.2 vs. 1.6±0.2, p = 0.001; mean±6S.D., fold induction at 12 h). CO postconditioning suppressed retinal p38 phosphorylation (p = 0.023 at 24 h) and induced the phosphorylation of ERK-1/2 (p,0.001 at 24 h). CO postconditioning inhibited the expression of HO-1. The activation of NF-κkB, microglia and Müller cells was potently inhibited by CO as well as immigration of proliferative microglia and macrophages into the retina. CO protected I/R-injured RGC with a therapeutic window at least up to 3 h (n = 8; RGC/mm²; mean±S.D.: 1255±327 I/R only vs. 1956±157 immediate CO treatment, vs. 1830±109 1.5 h time lag and vs. 1626±122 3 h time lag; p,0.001). Inhibition of ERK-1/2 did not counteract the CO effects (RGC/mm² : 1956±157 vs. 1931±124, mean±S.D., p = 0.799). Conclusion: Inhaled CO, administered after retinal ischemic injury, protects RGC through its strong anti-apoptotic and anti- inflammatory effects. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
4. Dobutamine mediates cytoprotection by induction of heat shock protein 70 in vitro.
- Author
-
Roesslein, Martin, Froehlich, Christian, Jans, Frank, Piegeler, Tobias, Goebel, Ulrich, and Loop, Torsten
- Subjects
- *
HEAT shock proteins , *DOBUTAMINE , *CYTOPROTECTION , *IN vitro studies , *PSYCHOLOGICAL stress , *NF-kappa B , *CELL death , *THERAPEUTICS - Abstract
Abstract: Aims: Dobutamine is cytoprotective when applied before a subsequent stress. However, the underlying molecular mechanism is unknown. Dobutamine also inhibits nuclear factor (NF)-?B in human T lymphocytes. Other inhibitors of NF-?B induce a so-called heat shock response. We hypothesized that dobutamine mediates protection from apoptotic cell death by the induction of a heat shock response. Main methods: Jurkat T lymphoma cells were preincubated with dobutamine (0.1, 0.5mM) before the induction of apoptosis (staurosporine, 2?M). DNA-binding of heat shock factor (HSF)-1 was analyzed by electrophoretic mobility shift assay, mRNA-expression of heat shock protein (hsp)70 and hsp90 by Northern Blot, activity of caspase-3 by fluorogenic caspase activity assay and cleavage of pro-caspase-3 by Western Blot. Apoptosis was assessed by flow cytometry after annexin V-fluorescein isothiocyanate staining. Hsp70 and hsp90 were inhibited using N-formyl-3,4-methylenedioxy-benzylidene-gamma-butyrolaetam and 17-allylamino-17-demethoxygeldana-mycin, respectively. All data are given as median and 25/75% percentile. Key findings: Pre-incubation with dobutamine inhibited staurosporine-induced annexin V-fluorescence (28 [20–32] % vs. 12 [9–15] % for dobutamine 0.1mM and 7 [5–12] % for dobutamine 0.5mM, p<0.001), cleavage of pro-caspase-3 as well as caspase-3-like activity (0.46 [0.40–0.48] vs. 0.32 [0.27–0.39] for Dobutamine 0.1mM and 0.20 [0.19–0.23] for Dobutamine 0.5mM, p<0.01). Dobutamine induced DNA-binding of HSF-1 and mRNA-expression of hsp70 and hsp90. While inhibition of Hsp90 had no effect, inhibition of Hsp70 increased the number of annexin V-positive cells (33 [32–36] % vs. 18 [16–24] %) and caspase-3-like activity (0.21 [0.19–0.23] vs. 0.16 [0.13–0.17], p<0.05). Significance: Dobutamine protects from apoptotic cell death via the induction of Hsp70. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.