Cargando…

β-Adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices

BACKGROUND: Inflammation acting in synergy with brain ischemia aggravates perinatal ischemic brain damage. The sensitizing effect of pro-inflammatory exposure prior to hypoxia is dependent on signaling by TNF-α through TNF receptor (TNFR) 1. Adrenoceptor (AR) activation is known to modulate the immu...

Descripción completa

Detalles Bibliográficos
Autores principales: Markus, Tina, Hansson, Stefan R, Cronberg, Tobias, Cilio, Corrado, Wieloch, Tadeusz, Ley, David
Formato: Texto
Lenguaje:English
Publicado: BioMed Central 2010
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3017519/
https://www.ncbi.nlm.nih.gov/pubmed/21172031
http://dx.doi.org/10.1186/1742-2094-7-94
_version_ 1782195906046590976
author Markus, Tina
Hansson, Stefan R
Cronberg, Tobias
Cilio, Corrado
Wieloch, Tadeusz
Ley, David
author_facet Markus, Tina
Hansson, Stefan R
Cronberg, Tobias
Cilio, Corrado
Wieloch, Tadeusz
Ley, David
author_sort Markus, Tina
collection PubMed
description BACKGROUND: Inflammation acting in synergy with brain ischemia aggravates perinatal ischemic brain damage. The sensitizing effect of pro-inflammatory exposure prior to hypoxia is dependent on signaling by TNF-α through TNF receptor (TNFR) 1. Adrenoceptor (AR) activation is known to modulate the immune response and synaptic transmission. The possible protective effect of [Formula: see text] and [Formula: see text] AR activation against neuronal damage caused by tissue ischemia and inflammation, acting in concert, was evaluated in murine hippocampal organotypic slices treated with lipopolysaccharide (LPS) and subsequently subjected to oxygen-glucose deprivation (OGD). METHOD: Hippocampal slices from mice were obtained at P6, and were grown in vitro for 9 days on nitrocellulose membranes. Slices were treated with β1(dobutamine)-, β2(terbutaline)-, α1(phenylephrine)- and α2(clonidine)-AR agonists (5 and 50 μM, respectively) during LPS (1 μg/mL, 24 h) -exposure followed by exposure to OGD (15 min) in a hypoxic chamber. Cell death in the slice CA1 region was assessed by propidium iodide staining of dead cells. RESULTS: Exposure to LPS + OGD caused extensive cell death from 4 up to 48 h after reoxygenation. Co-incubation with β1-agonist (50 μM) during LPS exposure before OGD conferred complete protection from cell death (P < 0.001) whereas the β2-agonist (50 μM) was partially protective (p < 0.01). Phenylephrine was weakly protective while no protection was attained by clonidine. Exposure to both β1- and β2-agonist during LPS exposure decreased the levels of secreted TNF-α, IL-6 and monocyte chemoattractant protein-1 and prevented microglia activation in the slices. Dobutamine remained neuroprotective in slices exposed to pure OGD as well as in TNFR1(-/- )and TNFR2(-/- )slices exposed to LPS followed by OGD. CONCLUSIONS: Our data demonstrate that activation of both β1- and β2-receptors is neuroprotective and may offer mechanistic insights valuable for development of neuro-protective strategies in neonates.
format Text
id pubmed-3017519
institution National Center for Biotechnology Information
language English
publishDate 2010
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-30175192011-01-10 β-Adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices Markus, Tina Hansson, Stefan R Cronberg, Tobias Cilio, Corrado Wieloch, Tadeusz Ley, David J Neuroinflammation Research BACKGROUND: Inflammation acting in synergy with brain ischemia aggravates perinatal ischemic brain damage. The sensitizing effect of pro-inflammatory exposure prior to hypoxia is dependent on signaling by TNF-α through TNF receptor (TNFR) 1. Adrenoceptor (AR) activation is known to modulate the immune response and synaptic transmission. The possible protective effect of [Formula: see text] and [Formula: see text] AR activation against neuronal damage caused by tissue ischemia and inflammation, acting in concert, was evaluated in murine hippocampal organotypic slices treated with lipopolysaccharide (LPS) and subsequently subjected to oxygen-glucose deprivation (OGD). METHOD: Hippocampal slices from mice were obtained at P6, and were grown in vitro for 9 days on nitrocellulose membranes. Slices were treated with β1(dobutamine)-, β2(terbutaline)-, α1(phenylephrine)- and α2(clonidine)-AR agonists (5 and 50 μM, respectively) during LPS (1 μg/mL, 24 h) -exposure followed by exposure to OGD (15 min) in a hypoxic chamber. Cell death in the slice CA1 region was assessed by propidium iodide staining of dead cells. RESULTS: Exposure to LPS + OGD caused extensive cell death from 4 up to 48 h after reoxygenation. Co-incubation with β1-agonist (50 μM) during LPS exposure before OGD conferred complete protection from cell death (P < 0.001) whereas the β2-agonist (50 μM) was partially protective (p < 0.01). Phenylephrine was weakly protective while no protection was attained by clonidine. Exposure to both β1- and β2-agonist during LPS exposure decreased the levels of secreted TNF-α, IL-6 and monocyte chemoattractant protein-1 and prevented microglia activation in the slices. Dobutamine remained neuroprotective in slices exposed to pure OGD as well as in TNFR1(-/- )and TNFR2(-/- )slices exposed to LPS followed by OGD. CONCLUSIONS: Our data demonstrate that activation of both β1- and β2-receptors is neuroprotective and may offer mechanistic insights valuable for development of neuro-protective strategies in neonates. BioMed Central 2010-12-20 /pmc/articles/PMC3017519/ /pubmed/21172031 http://dx.doi.org/10.1186/1742-2094-7-94 Text en Copyright ©2010 Markus et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (<url>http://creativecommons.org/licenses/by/2.0</url>), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research
Markus, Tina
Hansson, Stefan R
Cronberg, Tobias
Cilio, Corrado
Wieloch, Tadeusz
Ley, David
β-Adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices
title β-Adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices
title_full β-Adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices
title_fullStr β-Adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices
title_full_unstemmed β-Adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices
title_short β-Adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices
title_sort β-adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3017519/
https://www.ncbi.nlm.nih.gov/pubmed/21172031
http://dx.doi.org/10.1186/1742-2094-7-94
work_keys_str_mv AT markustina badrenoceptoractivationdepressesbraininflammationandisneuroprotectiveinlipopolysaccharideinducedsensitizationtooxygenglucosedeprivationinorganotypichippocampalslices
AT hanssonstefanr badrenoceptoractivationdepressesbraininflammationandisneuroprotectiveinlipopolysaccharideinducedsensitizationtooxygenglucosedeprivationinorganotypichippocampalslices
AT cronbergtobias badrenoceptoractivationdepressesbraininflammationandisneuroprotectiveinlipopolysaccharideinducedsensitizationtooxygenglucosedeprivationinorganotypichippocampalslices
AT ciliocorrado badrenoceptoractivationdepressesbraininflammationandisneuroprotectiveinlipopolysaccharideinducedsensitizationtooxygenglucosedeprivationinorganotypichippocampalslices
AT wielochtadeusz badrenoceptoractivationdepressesbraininflammationandisneuroprotectiveinlipopolysaccharideinducedsensitizationtooxygenglucosedeprivationinorganotypichippocampalslices
AT leydavid badrenoceptoractivationdepressesbraininflammationandisneuroprotectiveinlipopolysaccharideinducedsensitizationtooxygenglucosedeprivationinorganotypichippocampalslices