Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Hepatol ; 71(5): 930-941, 2019 11.
Article in English | MEDLINE | ID: mdl-31279900

ABSTRACT

BACKGROUND & AIMS: Cerebral oxidative stress plays an important role in the pathogenesis of hepatic encephalopathy (HE), but the underlying mechanisms are incompletely understood. Herein, we analyzed a role of heme oxygenase (HO)1, iron and NADPH oxidase 4 (Nox4) for the induction of oxidative stress and senescence in HE. METHODS: Gene and protein expression in human post-mortem brain samples was analyzed by gene array and western blot analysis. Mechanisms and functional consequences of HO1 upregulation were studied in NH4Cl-exposed astrocytes in vitro by western blot, qPCR and super-resolution microscopy. RESULTS: HO1 and the endoplasmic reticulum (ER) stress marker grp78 were upregulated, together with changes in the expression of multiple iron metabolism-related genes, in post-mortem brain samples from patients with liver cirrhosis and HE. NH4Cl elevated HO1 protein and mRNA in cultured astrocytes through glutamine synthetase (GS)-dependent upregulation of glutamine/fructose amidotransferases 1/2 (GFAT1/2), which blocked the transcription of the HO1-targeting miR326-3p in a O-GlcNAcylation dependent manner. Upregulation of HO1 by NH4Cl triggered ER stress and was associated with elevated levels of free ferrous iron and expression changes in iron metabolism-related genes, which were largely abolished after knockdown or inhibition of GS, GFAT1/2, HO1 or iron chelation. NH4Cl, glucosamine (GlcN) and inhibition of miR326-3p upregulated Nox4, while knockdown of Nox4, GS, GFAT1/2, HO1 or iron chelation prevented NH4Cl-induced RNA oxidation and astrocyte senescence. Elevated levels of grp78 and O-GlcNAcylated proteins were also found in brain samples from patients with liver cirrhosis and HE. CONCLUSION: The present study identified glucosamine synthesis-dependent protein O-GlcNAcylation as a novel mechanism in the pathogenesis of HE that triggers oxidative and ER stress, as well as senescence, through upregulation of HO1 and Nox4. LAY SUMMARY: Patients with liver cirrhosis frequently exhibit hyperammonemia and suffer from cognitive and motoric dysfunctions, which at least in part involve premature ageing of the astrocytes in the brain. This study identifies glucosamine and an O-GlcNAcylation-dependent disruption of iron homeostasis as novel triggers of oxidative stress, thereby mediating ammonia toxicity in the brain.


Subject(s)
Ammonia/pharmacology , Cellular Senescence/genetics , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1/metabolism , Hepatic Encephalopathy/metabolism , Membrane Proteins/metabolism , Oxidative Stress/drug effects , Protein Processing, Post-Translational/drug effects , Up-Regulation/genetics , Adult , Aged , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Female , Glucosamine/biosynthesis , Heme Oxygenase-1/genetics , Hepatic Encephalopathy/etiology , Humans , Liver Cirrhosis/complications , Male , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Oxidative Stress/genetics , Rats , Rats, Wistar , Temporal Lobe/metabolism , Temporal Lobe/pathology
2.
Sci Rep ; 7(1): 7938, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28801579

ABSTRACT

The devastating consequences of hepatic failure include hepatic encephalopathy, a severe, life threatening impairment of neuronal function. Hepatic encephalopathy is caused by impaired hepatic clearance of NH4+. Cellular NH4+ uptake is accomplished mainly by the Na+,K+,2Cl- cotransporter. Here we show that hepatic clearance of NH4+ is impaired in TNFα deficient as well as TNFR1&TNFR2 double knockout mice, which both develop hyperammonemia. Despite impaired hepatic clearance of NH4+, TNFα deficient mice and TNFR1 deficient mice were protected against acute ammonia intoxication. While 54% of the wild-type mice and 60% of TNFR2 deficient mice survived an NH4+ load, virtually all TNFα deficient mice and TNFR1 deficient mice survived the treatment. Conversely, TNFα treatment of wild type mice sensitized the animals to the toxic effects of an NH4+ load. The protection of TNFα-deficient mice against an NH4+ load was paralleled by decreased cerebral expression of NKCC1. According to the present observations, inhibition of TNFα formation and/or NKCC1 may be strategies to favorably influence the clinical course of hepatic encephalopathy.


Subject(s)
Hyperammonemia/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Solute Carrier Family 12, Member 2/metabolism , Tumor Necrosis Factor-alpha/deficiency , Ammonia/toxicity , Animals , Brain/metabolism , Hyperammonemia/metabolism , Liver/chemistry , Mice , Mice, Knockout , Solute Carrier Family 12, Member 2/genetics , Tumor Necrosis Factor-alpha/genetics , Up-Regulation
3.
Sci Rep ; 6: 34946, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27721451

ABSTRACT

To date, unequivocal neuroanatomical features have been demonstrated neither for sporadic nor for familial schizophrenia. Here, we investigated the neuroanatomical changes in a transgenic rat model for a subset of sporadic chronic mental illness (CMI), which modestly overexpresses human full-length, non-mutant Disrupted-in-Schizophrenia 1 (DISC1), and for which aberrant dopamine homeostasis consistent with some schizophrenia phenotypes has previously been reported. Neuroanatomical analysis revealed a reduced density of dopaminergic neurons in the substantia nigra and reduced dopaminergic fibres in the striatum. Parvalbumin-positive interneuron occurrence in the somatosensory cortex was shifted from layers II/III to V/VI, and the number of calbindin-positive interneurons was slightly decreased. Reduced corpus callosum thickness confirmed trend-level observations from in vivo MRI and voxel-wise tensor based morphometry. These neuroanatomical changes help explain functional phenotypes of this animal model, some of which resemble changes observed in human schizophrenia post mortem brain tissues. Our findings also demonstrate how a single molecular factor, DISC1 overexpression or misassembly, can account for a variety of seemingly unrelated morphological phenotypes and thus provides a possible unifying explanation for similar findings observed in sporadic schizophrenia patients. Our anatomical investigation of a defined model for sporadic mental illness enables a clearer definition of neuroanatomical changes associated with subsets of human sporadic schizophrenia.


Subject(s)
Brain/pathology , Dopaminergic Neurons/physiology , Interneurons/physiology , Nerve Tissue Proteins/biosynthesis , Parvalbumins/analysis , Schizophrenia/physiopathology , Animals , Biometry , Disease Models, Animal , Gene Expression , Histocytochemistry , Humans , Magnetic Resonance Imaging , Male , Rats, Sprague-Dawley , Rats, Transgenic
4.
Sci Rep ; 6: 18493, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26755400

ABSTRACT

Hepatic encephalopathy is a neuropsychiatric syndrome evolving from cerebral osmotic disturbances and oxidative/nitrosative stress. Ammonia, the main toxin of hepatic encephalopathy, triggers astrocyte senescence in an oxidative stress-dependent way. As miRNAs are critically involved in cell cycle regulation and their expression may be regulated by oxidative stress, we analysed, whether astrocyte senescence is a consequence of ammonia-induced miRNA expression changes. Using a combined miRNA and gene microarray approach, 43 miRNA species which were downregulated and 142 genes which were upregulated by NH4Cl (5 mmol/l, 48 h) in cultured rat astrocytes were found. Ammonia-induced miRNA and gene expression changes were validated by qPCR and 43 potential miRNA target genes, including HO-1, were identified by matching upregulated mRNA species with predicted targets of miRNA species downregulated by ammonia. Inhibition of HO-1 targeting miRNAs which were downregulated by NH4Cl strongly upregulated HO-1 mRNA and protein levels and inhibited astrocyte proliferation in a HO-1-dependent way. Preventing ammonia-induced upregulation of HO-1 by taurine (5 mmol/l) as well as blocking HO-1 activity by tin-protoporphyrine IX fully prevented ammonia-induced proliferation inhibition and senescence. The data suggest that ammonia induces astrocyte senescence through NADPH oxidase-dependent downregulation of HO-1 targeting miRNAs and concomitant upregulation of HO-1 at both mRNA and protein level.


Subject(s)
Ammonia/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Gene Expression Regulation/drug effects , MicroRNAs/genetics , Animals , Cell Proliferation , Cells, Cultured , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cluster Analysis , Gene Expression Profiling , Glutamine/biosynthesis , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , NADPH Oxidases/metabolism , Rats , Reproducibility of Results
5.
Proc Natl Acad Sci U S A ; 112(17): 5521-6, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25870278

ABSTRACT

Urea cycle defects and acute or chronic liver failure are linked to systemic hyperammonemia and often result in cerebral dysfunction and encephalopathy. Although an important role of the liver in ammonia metabolism is widely accepted, the role of ammonia metabolizing pathways in the liver for maintenance of whole-body ammonia homeostasis in vivo remains ill-defined. Here, we show by generation of liver-specific Gln synthetase (GS)-deficient mice that GS in the liver is critically involved in systemic ammonia homeostasis in vivo. Hepatic deletion of GS triggered systemic hyperammonemia, which was associated with cerebral oxidative stress as indicated by increased levels of oxidized RNA and enhanced protein Tyr nitration. Liver-specific GS-deficient mice showed increased locomotion, impaired fear memory, and a slightly reduced life span. In conclusion, the present observations highlight the importance of hepatic GS for maintenance of ammonia homeostasis and establish the liver-specific GS KO mouse as a model with which to study effects of chronic hyperammonemia.


Subject(s)
Glutamate-Ammonia Ligase/metabolism , Hyperammonemia/enzymology , Liver/enzymology , Animals , Behavior, Animal , Brain/metabolism , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Gene Targeting , Glutamate-Ammonia Ligase/genetics , Hyperammonemia/genetics , Hyperammonemia/pathology , Hyperammonemia/physiopathology , Liver/metabolism , Liver/physiopathology , Locomotion , Memory , Mice , Mice, Knockout , Oxidative Stress/genetics
6.
Glia ; 63(1): 37-50, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25092802

ABSTRACT

Hepatic encephalopathy (HE) is a frequent complication of liver cirrhosis and is due to a low-grade cerebral edema associated with oxidative/nitrosative stress. Recent reports suggest that cognitive impairment in cirrhotic patients may not resolve completely after an attack of manifest HE. As astrocyte dysfunction is central to the pathogenesis of HE and astrocytes are critically involved in synaptic plasticity, we tested for sustained impairment of astrocyte function by analyzing expression levels of senescence biomarkers in ammonia-treated cultured rat astrocytes and in postmortem brain samples from cirrhotic patients with or without HE. NH4 Cl time- and dose-dependently inhibited proliferation of cultured astrocytes by up to 45% (5 mmol/L, 72 h) and strongly increased senescence-associated ß-galactosidase activity. Inhibition of astrocyte proliferation by ammonia was mediated by a l-methionine sulfoximine-, oxidative stress-, and p38(MAPK) -dependent activation of p53 associated with enhanced transcription of cell cycle inhibitory genes GADD45α and p21. Mitochondria and the nucleus were identified as sources of oxygen radical formation after prolonged NH4 Cl exposure. Concurrently, NH4 Cl (5 mmol/L) treatment inhibited both epidermal growth factor- and brain-derived neurotrophic factor (BDNF)-induced proliferation as well as BDNF-mediated astrocyte morphology changes through downregulation of the respective growth factor receptors epidermal growth factor receptor and truncated tyrosine receptor kinase B. Increased mRNA expression levels of senescence-associated genes were also found in post mortem brain samples from patients with liver cirrhosis with HE, but not in those without HE. The data suggest that ammonia toxicity and HE are associated with premature astrocyte senescence, which may impair neurotransmission and contribute to persistence of cognitive disturbances after resolution of episodes of overt HE.


Subject(s)
Ammonia/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Cellular Senescence/drug effects , Cerebral Cortex/drug effects , Hepatic Encephalopathy/drug therapy , Hepatic Encephalopathy/metabolism , Ammonia/metabolism , Animals , Brain Edema/etiology , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Rats, Wistar , Reactive Oxygen Species/metabolism
7.
Brain Res ; 1570: 54-60, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24835408

ABSTRACT

The purpose of the present study was to determine whether a unilateral photothrombotic brain lesion induces bilateral ischemic tolerance towards a subsequent severe ischemia performed 5 days later. Severe ischemia was induced by transient (1h; t) or permanent (p) occlusion of the middle cerebral artery (MCAO). Rats were sacrificed 24h later. Preconditioning reduced the size of subsequent infarcts in both hemispheres. This effect was most prominent with tMCAO, and ipsilateral preconditioning was more effective than contralateral preconditioning (% of hemispheric volume, mean ± SD: 31.9 ± 3.7 to 19.0 ± 10.3 with ipsilateral tMCAO; 31.9 ± 3.7 to 22.9 ± 4.9 with contralateral tMCAO; 64.7 ± 4.3% to 47.2 ± 12.5% with ipsilateral pMCAO; 64.7 ± 4.3% to 53.1 ± 8.9% with contralateral pMCAO). Ischemic preconditioning was associated with a successive bilateral up-regulation of superoxide dismutases which may be involved in the development of ischemic tolerance. Our data suggest that a focal ischemic brain lesion induces neuroprotective mechanisms in extensive brain areas and thus cause bilateral ischemic tolerance within a certain time window.


Subject(s)
Brain Ischemia/physiopathology , Brain/physiopathology , Functional Laterality/physiology , Infarction, Middle Cerebral Artery/physiopathology , Ischemic Preconditioning/methods , Animals , Brain/pathology , Brain Ischemia/pathology , Disease Models, Animal , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Male , Photic Stimulation , Random Allocation , Rats, Wistar , Severity of Illness Index , Superoxide Dismutase/metabolism , Thrombosis , Up-Regulation/physiology , Weight Loss
8.
Epilepsia ; 53(11): 1987-97, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22957946

ABSTRACT

PURPOSE: A disturbed balance between excitatory and inhibitory neurotransmission underlies epileptic activity, although reports concerning neurotransmitter systems involved remain controversial. METHODS: We quantified densities of 15 receptors in neocortical biopsies from patients with pharmacoresistant focal temporal lobe epilepsy and autopsy controls, and searched for correlations between density alterations and clinical factors or the occurrence of spontaneous synaptic potentials in vitro. KEY FINDINGS: α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), kainate, N-methyl-d-aspartate (NMDA), peripheral benzodiazepine, muscarinic (M)(1) , M(2) , nicotinic, α(1) , α(2h) , serotonin (5-HT)(1A) , and adenosine (A)(1) receptor densities were significantly altered in biopsies. The epileptic cohort was subdivided based on clinical (febrile seizures, hippocampal sclerosis, neocortical pathologies, surgery outcome) or electrophysiologic (spontaneous field potentials) criteria, resulting in different patterns of significantly altered receptor types when comparing a given epileptic group with controls. Only AMPA, kainate, M(2) , and 5-HT(1A) receptors were always significantly altered. γ-Aminobutyric acid (GABA)(A) , GABA(B) , and 5-HT(2) receptor alterations were never significant. Correlation patterns between receptor alterations and illness duration or seizure frequency varied depending on whether the epileptic cohort was considered as a whole or subdivided. SIGNIFICANCE: Neocortical temporal lobe epilepsy is associated with a generalized receptor imbalance resulting in a net potentiation of excitatory neurotransmission. Peripheral benzodiazepine receptor alterations highlight that astrocytes are also impaired by seizure activity.


Subject(s)
Epilepsies, Partial/metabolism , Neocortex/metabolism , Receptors, Cell Surface/metabolism , Adult , Aged , Cohort Studies , Electroencephalography/methods , Epilepsies, Partial/physiopathology , Female , Humans , Ligands , Male , Middle Aged , Protein Binding/physiology , Young Adult
9.
Biol Chem ; 392(12): 1155-65, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22050230

ABSTRACT

Ammonia is a major player in the pathogenesis of hepatic encephalopathy (HE) and affects astrocyte function by triggering a self-amplifying cycle between osmotic and oxidative stress. We recently demonstrated that hypoosmotic astrocyte swelling rapidly stimulates nitric oxide (NO) production and increases intracellular free Zn(2+) concentration ([Zn(2+)](i)). Here we report effects of ammonia on [Zn(2+)](i) homeostasis and NO synthesis. In cultured rat astrocytes, NH(4)Cl (5 mm) increased within 6 h both cytosolic and mitochondrial [Zn(2+)]. The [Zn(2+)](i) increase was transient and was mimicked by the nonmetabolizable CH(3)NH(3)Cl, and it was dependent on NO formation, as evidenced by the sensitivity toward the nitric oxide synthase inhibitor N(G)-monomethyl-l-arginine. The NH(4)Cl-induced NO formation was sensitive to the Ca(2+) chelator 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetra(acetoxymethyl) ester and increases in both NO and [Zn(2+)](i) were blocked by the N-methyl-d-aspartate receptor antagonist MK-801. The NH(4)Cl-triggered increase in [Zn(2+)](i) was followed by a Zn(2+)-dependent nuclear appearance of the metal response element-binding transcription factor and metallothionein messenger RNA (mRNA) induction. Metallothionein mRNA was also increased in vivo in rat cerebral cortex 6 h after an NH(4)Ac challenge. NH(4)Cl increased peripheral-type benzodiazepine receptor (PBR) protein expression, whereas PBR mRNA levels were decreased in a Zn(2+)-independent manner. The Zn(2+)-dependent upregulation of metallothionein following ammonia intoxication may reflect a cytoprotective response, whereas the increase in PBR expression may augment HE development.


Subject(s)
Ammonia/pharmacology , Astrocytes/drug effects , Metallothionein/genetics , Nitric Oxide/metabolism , Up-Regulation/drug effects , Zinc/metabolism , Animals , Astrocytes/metabolism , Cells, Cultured , Male , RNA, Messenger/genetics , Rats , Rats, Wistar
10.
Glia ; 58(15): 1794-805, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20665558

ABSTRACT

TGR5 (Gpbar-1) is a membrane-bound bile acid receptor in the gastrointestinal tract and immune cells with pleiotropic actions. As shown in the present study, TGR5 is also expressed in astrocytes and neurons. Here, TGR5 may act as a neurosteroid receptor, which is activated by nanomolar concentrations of 5ß-pregnan-3α-ol-20-one and micromolar concentrations of 5ß-pregnan-3α-17α-21-triol-20-one and 5α-pregnan-3α-ol-20-one (allopregnanolone). TGR5 stimulation in astrocytes and neurons is coupled to adenylate cyclase activation, elevation of intracellular Ca(2+) and the generation of reactive oxygen species. In cultured rat astrocytes, TGR5 mRNA is downregulated in the presence of neurosteroids and ammonia already at concentrations of 0.5 mmol L(-1). Furthermore, TGR5 protein levels are significantly reduced in isolated rat astrocytes after incubation with ammonia. A marked downregulation of TGR5 mRNA is also found in cerebral cortex from cirrhotic patients dying with hepatic encephalopathy (HE) when compared with brains from noncirrhotic control subjects. It is concluded that TGR5 is a novel neurosteroid receptor in brain with implications for the pathogenesis of HE.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Gene Expression Regulation/physiology , Neurons/metabolism , Receptors, G-Protein-Coupled/physiology , Animals , Animals, Newborn , Astrocytes/drug effects , Brain/cytology , CREB-Binding Protein/metabolism , Calcium/metabolism , Cells, Cultured , Cholagogues and Choleretics/pharmacology , Coculture Techniques , Colforsin/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Humans , Luminescent Proteins , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurotransmitter Agents/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Statistics, Nonparametric , Taurolithocholic Acid/pharmacology , Transfection/methods
11.
Epilepsia ; 49(10): 1733-48, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18479397

ABSTRACT

PURPOSE: The astrocyte-specific glutamine synthetase (GS) plays a key role in glutamate recycling and Gamma-aminobutyric acid (GABA) metabolism. Changes in the expression or activity of GS have been proposed to contribute to epileptogenesis. The mechanisms or how and where GS may contribute to epilepsy is still a matter of discussion. Here we asked the question whether brain regions, which show an astrocytic stress response respond with alterations of GS. METHODS: Biochemical and histological alterations of GS, HSP-27, and GFAP were studied after pentylenetetrazole-induced repetitive epileptic seizures (PIRS) in rats using a topographical quantification of the GS-immunoreactivity (GSIR) in relation to the focal heat shock response (HSR). Saline-treated rats served as controls and rats treated by the GS-inhibitor, L-methionine-sulfoximine (MSO) served as a positive control. RESULTS: No changes in the amount of GSIR and GS-protein occurred during PIRS. A significant reduction of GSIR was observed by histochemistry (in situ) and in native (nonheated) protein extracts of MSO-treated rats. In rats affected by PIRS, GS-activity showed a significant, region-specific reduction in association with a nitration of the enzyme. DISCUSSION: These results show that neither PIRS nor GS-inhibition reduced the amount of GS protein, but that MSO interferes with antibody binding to native GS. PIRS resulted in a focal increase of astrocytic stress response, whereas MSO caused a widespread, homogeneous astrocytic HSR independent from quantitative changes of GS content. In rats with PIRS the regions showing a strong glial HSR, respond with reduced GS-activity and GS-nitration, which all together are clear indicators of a nitrosative stress response.


Subject(s)
Epilepsy/chemically induced , Epilepsy/metabolism , Epilepsy/physiopathology , Glutamate-Ammonia Ligase/metabolism , Pentylenetetrazole , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Disease Models, Animal , Epilepsy/pathology , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , HSP27 Heat-Shock Proteins/metabolism , Immunoprecipitation/methods , Male , Models, Biological , Rats , Rats, Wistar
12.
Hepatology ; 48(2): 567-79, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18506841

ABSTRACT

UNLABELLED: Oxidative stress plays a major role in cerebral ammonia toxicity and the pathogenesis of hepatic encephalopathy (HE). As shown in this study, ammonia induces a rapid RNA oxidation in cultured rat astrocytes, vital mouse brain slices, and rat brain in vivo. Ammonia-induced RNA oxidation in cultured astrocytes is reversible and sensitive to MK-801, 1,2-Bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid, apocynin, epigallocatechin gallate, and polyphenon 60, suggesting the involvement of N-methyl-D-aspartic acid (NMDA) receptor activation, Ca(2+), nicotinamide adenine dinucleotide phosphate, and reduced form (NADPH) oxidase-dependent oxidative stress. Also, hypo-osmolarity, tumor necrosis factor alpha (TNF-alpha), and diazepam increase RNA oxidation in cultured astrocytes, suggesting that the action of different HE-precipitating factors converges at the level of RNA oxidation. Among the oxidized RNA species, 18S-rRNA and the messenger RNA (mRNA) coding for the glutamate/aspartate transporter (GLAST) were identified. Cerebral RNA oxidation in acutely ammonia-loaded rats in vivo is reversible and predominates in neuronal soma and perivascular astrocyte processes. In neuronal dendrites, oxidized RNA colocalizes with the RNA-binding splicing protein neurooncological ventral antigen (NOVA)-2 within putative RNA transport granules, which are also found in close vicinity to postsynaptic spines. This indicates that oxidized RNA species may participate in postsynaptic protein synthesis, which is a biochemical substrate for learning and memory consolidation. Neuronal and astroglial RNA oxidation increases also in vital mouse brain slices treated with ammonia and TNF-alpha, respectively. CONCLUSION: Cerebral RNA oxidation is identified as a not yet recognized consequence of acute ammonia intoxication. RNA oxidation may affect gene expression and local protein synthesis and thereby provide another link between reactive oxygen species (ROS)/reactive nitrogen oxide species (RNOS) production and ammonia toxicity.


Subject(s)
Ammonia/pharmacology , Astrocytes/metabolism , Brain/metabolism , RNA/metabolism , Amino Acid Transport System X-AG/genetics , Animals , Astrocytes/drug effects , Brain/cytology , Cells, Cultured , Dendrites/metabolism , Diazepam/pharmacology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Osmolar Concentration , Oxidation-Reduction/drug effects , RNA Transport , RNA, Messenger/metabolism , RNA, Ribosomal, 18S/metabolism , Rats , Rats, Wistar , Tissue Distribution , Tumor Necrosis Factor-alpha/pharmacology
13.
Glia ; 55(7): 758-71, 2007 May.
Article in English | MEDLINE | ID: mdl-17352382

ABSTRACT

The role of NADPH oxidase (NOX) and the regulatory subunit p47(phox) for hypoosmotic ROS generation was studied in cultured rat astrocytes and brain slices of wilde type and p47(phox) knock-out mice. Cultured rat astrocytes express mRNAs encoding for the regulatory subunit p47(phox), NOX1, 2, and 4, and the dual oxidases (DUOX)1 and 2, but not NOX3. Hypoosmotic (205 mosmol/L) swelling of cultured astrocytes induced a rapid generation of ROS that was accompanied by serine phosphorylation of p47(phox) and prevented by the NADPH oxidase inhibitor apocynin. Apocynin also impaired the hypoosmotic tyrosine phosphorylation of Src. Both, hypoosmotic ROS generation and p47(phox) serine phosphorylation were sensitive to the acidic sphingomyelinase inhibitors AY9944 and desipramine, the protein kinase C (PKC)zeta-inhibitory pseudosubstrate peptide, the NMDA receptor antagonist MK-801 and the intracellular Ca(2+) chelator BAPTA-AM. Also hypoosmotic exposure of wilde type mouse cortical brain slices increased ROS generation, which was allocated in part to the astrocytes and which was absent in presence of apocynin and in cortical brain slices from p47(phox) knock-out mice. Also ammonia induced a rapid ROS production in cultured astrocytes and brain slices, which was sensitive to apocynin. The data suggest that astrocyte swelling triggers a p47(phox)-dependent NADPH oxidase-catalyzed ROS production. The findings further support a close interrelation between osmotic and oxidative stress in astrocytes, which may be relevant to different brain pathologies including hepatic encephalopathy.


Subject(s)
Brain Edema/metabolism , Hepatic Encephalopathy/metabolism , Hyperammonemia/metabolism , NADPH Oxidases/metabolism , Oxidative Stress , Ammonia/metabolism , Ammonia/toxicity , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Brain Edema/physiopathology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Hepatic Encephalopathy/physiopathology , Hyperammonemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/genetics , Organ Culture Techniques , Osmotic Pressure , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Water-Electrolyte Balance/drug effects , Water-Electrolyte Balance/physiology
14.
Cell Tissue Res ; 326(3): 707-14, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16819625

ABSTRACT

Nitric-oxide-sensitive guanylyl cyclase (NO-sGC) plays a pivotal role in many second messenger cascades. Neurotransmission- and neuropathology-related changes in NO-sGC have been suggested. However, the cellular localization of NO-sGC in primate brains, including humans, remains unknown. Biochemical evidence has linked the alpha(2)-subunit of NO-sGC directly to neurotransmission in rodents. Here, we have used a recently characterized subunit-specific antibody for the localization of the alpha(2)-subunit on sections from the cerebelli of the common marmoset (Callithrix jacchus; New World monkey) and macaque monkeys (Macaca mulatta, M. fascicularis; Old World monkeys). In contrast to the more ubiquitous cytoplasmic presence of subunit-beta(1), the alpha(2)-subunit is mainly confined to the somato-dendritic membrane including the spines of the Purkinje cells. Only limited colocalization with presynaptically localized synaptophysin has been seen under our staining conditions, indicating a higher abundance of subunit-alpha(2) at the postsynaptic site. This localization indicates that subunit-alpha(2) links NO-sGC to neurotransmission, whereas subunit-beta(1) may act as a cytoplasmic regulator/activator by contributing to active heterodimer formation via translocation from the cytoplasm to the cell membrane. The last-mentioned action may be a prerequisite for generating nitric-oxide-dependent, subcellular, and postsynaptically localized cGMP signals along neuronal processes.


Subject(s)
Cerebellum/enzymology , Cerebellum/metabolism , Guanylate Cyclase/metabolism , Primates/metabolism , Protein Subunits/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Antibodies, Monoclonal/metabolism , Calbindins , Callithrix , Cerebellum/cytology , Cyclic GMP/physiology , Guanylate Cyclase/genetics , Immunohistochemistry , Macaca fascicularis , Macaca mulatta , Male , Protein Subunits/genetics , Purkinje Cells/enzymology , Receptors, Cytoplasmic and Nuclear/genetics , S100 Calcium Binding Protein G/metabolism , Signal Transduction/physiology , Soluble Guanylyl Cyclase , Species Specificity
15.
Arch Biochem Biophys ; 449(1-2): 104-14, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16579953

ABSTRACT

Protein tyrosine nitration may be relevant for the pathogenesis of hepatic encephalopathy (HE). Infections, sepsis, and trauma precipitate HE episodes. Recently, serum levels of tumor necrosis factor (TNF)-alpha were shown to correlate with severity of HE in chronic liver failure. Here the effects of inflammatory cytokines on protein tyrosine nitration in cultured rat astrocytes and rat brain in vivo were studied. In cultured rat astrocytes TNF-alpha (50 pg/ml-10 ng/ml) within 6h increased protein tyrosine nitration. TNF-alpha-induced tyrosine nitration was related to an increased formation of reactive oxygen and nitrogen intermediates, which was downstream from a NMDA-receptor-dependent increase of intracellular [Ca(2+)](i) and nNOS-catalyzed NO production. Astroglial tyrosine nitration was also elevated in brains of rats receiving a non-lethal injection of lipopolysaccharide, as indicated by colocalization of nitrotyrosine immunoreactivity with glial fibrillary acidic protein and glutamine synthetase, and by identification of the glutamine synthetase among the tyrosine-nitrated proteins. It is concluded that reactive oxygen and nitrogen intermediates as well as protein tyrosine nitration by inflammatory cytokines may alter astrocyte function in an NMDA-receptor-, Ca(2+)-, and NOS-dependent fashion. This may be relevant for the pathogenesis of HE and other conditions involving cytokine exposure the brain.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Cytokines/administration & dosage , Inflammation Mediators/administration & dosage , Nerve Tissue Proteins/metabolism , Tyrosine/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Brain/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Lipopolysaccharides/administration & dosage , Rats , Rats, Wistar
16.
Anat Embryol (Berl) ; 211(2): 129-37, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16502013

ABSTRACT

The inducible prostaglandin synthase, cyclooxygenase-2, is upregulated in response to cerebral ischemia and contributes to potentiation of oxidative injury. Cyclooxygenase-2 expression is regulated by retinoic acid receptors, which form heterodimers with vitamin D receptors and vitamin D. In addition, vitamin D has been reported to have neuroprotective qualities. The aim of this study was to examine whether the biologically active vitamin D3-metabolite 1alpha,25-dihydroxyvitamin D3 (1,25-D3), influences the expression of inducible cyclooxygenase-2 in photothrombotically lesioned brain or is part of an independent neuroprotective mechanism. We compared groups of nonlesioned control rats and infarcted animals, which were treated with either 1,25-D3 or solvent at different times postlesion. In control animals, cyclooxygenase-2 immunoreactivity was readily evident in almost all cortical neurons of layers II/III as well as in a few pyramidal cells in layer V. Following photothrombotic infarction of the right cortical hindlimb area, there was a significant, but transient, increase in cyclooxygenase-2 labeling which was restricted to neurons of the injured hemisphere in both 1,25- D3-treated and solvent-treated rats. Highest levels of cyclooxygenase-2 immunoreactivity were seen at 12 and 24 h postlesion, followed by a gradual decrease at later time points. However, no significant differences were detected between 1,25-D3-treated and solvent-treated lesioned rats, indicating that postischemic neuronal cyclooxygenase-2 upregulation is not influenced by 1,25-D3. It is concluded that the neuroprotective effect of 1,25-D3 does not depend on modulations of neuronal COX-2 expression caused by postlesional hyperexcitation.


Subject(s)
Calcitriol/pharmacology , Cerebral Cortex/enzymology , Cyclooxygenase 2/metabolism , Intracranial Thrombosis/drug therapy , Intracranial Thrombosis/enzymology , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Disease Models, Animal , Immunoenzyme Techniques , Intracranial Thrombosis/pathology , Male , Rats , Rats, Wistar
17.
J Chem Neuroanat ; 30(1): 1-16, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15921884

ABSTRACT

Heat shock protein-27 (HSP-27) is an inducible stress response protein. It inhibits apoptotic cell death and is a reliable marker for oxidative stress. We studied the induction of HSP-27 in rat brains on days 1, 4 and 14 after repeated, pentylenetetrazole (PTZ)-induced seizures using immunohistochemisty. Saline treated control rats showed no induction of HSP-27. HSP-27 reactive astrocytes were rarely seen 1 or 4 days after PTZ injection. When present, single astrocytes were located in the cortex and/or the hippocampus. After 14 days PTZ treatment, a bilateral distribution of HSP-27 immunoreactive glia was present in piriform and entorhinal cortices and in the dentate gyrus of most brains. Rats with most intense HSP-27 upregulation showed HSP-27 in amygdala and thalamic nuclei. Astrocytes associated with blood vessels presented strongest HSP-27 staining, but did not show upregulation of gial fibrillary acidic protein and none responded with HSP-47 expression. Additionally, HSP-27 immunoreactivity increased in the endothelial cells of blood vessels in the affected brain regions, although no neuronal induction occurred. Contrastingly, a subconvulsive dose of the glutamine synthetase inhibitor L-methionine sulfoxime, which acts directly on astrocytes, resulted in a rapid, homogeneous astrocyte-specific HSP-27 upregulation within 24 h. Thus, repeated PTZ-induced seizure activity elicits a focal "heat shock" response in endothelial cells and astrocytes of selected cerebral regions indicating that expression of HSP-27 occurred in a seizure-dependent manner within the affected cerebral circuitries. Therefore, this PTZ-model of repeated seizure activity exhibited a cortical pattern of HSP-27 expression which is most comparable to that known from patients with epilepsy.


Subject(s)
Astrocytes/metabolism , Brain/blood supply , Brain/metabolism , Heat-Shock Proteins/biosynthesis , Animals , Astrocytes/drug effects , Blotting, Western , Convulsants/toxicity , Disease Models, Animal , Electroencephalography , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Epilepsy/chemically induced , Functional Laterality , Glial Fibrillary Acidic Protein/metabolism , Glutamate-Ammonia Ligase/antagonists & inhibitors , Immunohistochemistry , Male , Pentylenetetrazole/toxicity , Rats , Rats, Wistar , Time Factors , Up-Regulation
18.
Arch Biochem Biophys ; 439(1): 70-9, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15922286

ABSTRACT

1alpha,25-(OH)(2)-vitamin-D(3) (1,25-D(3)) and 17beta-estradiol are both known to act neuroprotective in certain experimental in vitro and in vivo settings. We studied the effects of 1,25-D(3) or 17beta-estradiol or their combined application on heat shock protein-27 (HSP-27) distribution after focal cortical ischemia using the photothrombosis model. HSP-27 is a well-established marker of the cerebral oxidative stress response and a potent inhibitor of apoptosis. Lesioned rats were injected i.p. one hour after injury with either 1 microg 1,25-D(3)/kg or 7 microg 17beta-estradiol/kg or a combination of both steroids. Groups of non-lesioned steroid-treated rats and lesioned, solvent-treated rats served as controls. Treatment with both steroids did not affect the size of the lesion. In addition, 17beta-estradiol resulted in significant reduction of HSP-27 induction, whereas the combination of 1,25-D(3)+17beta-estradiol resulted in a highly significant reduction of HSP-27 within the infracted cerebral cortex, indicating that both steroids act synergistically in a protective manner.


Subject(s)
Brain Ischemia/metabolism , Cerebral Cortex/metabolism , Estradiol/administration & dosage , Gene Expression Regulation, Enzymologic/drug effects , Heat-Shock Proteins/biosynthesis , Vitamin D/analogs & derivatives , Vitamin D/administration & dosage , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Brain Ischemia/pathology , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Drug Synergism , Male , Oxidative Stress/drug effects , Rats , Rats, Wistar
19.
Epilepsia ; 45(12): 1549-59, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15571513

ABSTRACT

PURPOSE: Heat shock protein-27 (HSP-27) belongs to the group of small heat shock proteins that become induced in response to various pathologic conditions. HSP-27 has been shown to protect cells and subcellular structures, particularly mitochondria, and serves as a carrier for estradiol. It is a reliable marker for tissues affected by oxidative stress. Oxidative stress and related cellular defence mechanisms are currently thought to play a major role during experimentally induced epileptic neuropathology. We addressed the question whether HSP-27 becomes induced in the neocortex resected from patients with pharmacoresistant epilepsy. METHODS: Human epileptic temporal neocortex was obtained during neurosurgery, and control tissue was obtained at autopsy from subjects without known neurologic diseases. The tissues were either frozen for Western blot analysis or fixed in Zamboni's fixative for the topographic detection of HSP-27 at the cellular level by means of immunohistochemistry. RESULTS: HSP-27 was highly expressed in all epilepsy specimens and in the cortex of a patient who died in the final stage of multiple sclerosis (positive control), whereas only low amounts of HSP-27 were detectable in control brains. In epilepsy patients, HSP-27 was present in astrocytes and in the walls of blood vessels. The intracortical distribution patterns varied strongly among the epilepsy specimens. CONCLUSIONS: These results demonstrate that HSP-27 becomes induced in response to epileptic pathology. Although the functional aspects of HSP-27 induction during human epilepsy have yet to be elucidated, it can be concluded that HSP-27 is a marker for cortical regions in which a stress response has been caused by seizures.


Subject(s)
Epilepsy/metabolism , Heat-Shock Proteins/metabolism , Temporal Lobe/chemistry , Adolescent , Adult , Aged , Astrocytes/chemistry , Astrocytes/metabolism , Biomarkers , Blood-Brain Barrier/metabolism , Blotting, Western , Child, Preschool , Epilepsy/diagnosis , Epilepsy/surgery , Female , HSP27 Heat-Shock Proteins , Heat-Shock Proteins/analysis , Humans , Immunohistochemistry , Male , Middle Aged , Molecular Chaperones , Neocortex/chemistry , Neocortex/metabolism , Neoplasm Proteins/analysis , Neoplasm Proteins/metabolism , Temporal Lobe/metabolism , Temporal Lobe/surgery , Up-Regulation
20.
J Chem Neuroanat ; 28(4): 225-38, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15531134

ABSTRACT

In ischemic cerebral injuries a cascade of degenerative mechanisms, all participating in the development of oxidative stress, influence the condition of the tissue. The survival of viable tissue affected by secondary injury largely depends on the balance between endogenous protective mechanisms and the ongoing degenerative processes. The inducible enzyme, heme oxygenase-1 metabolizes and thus detoxifies free heme to the powerful endogenous antioxidants biliverdin and bilirubin therefore enhancing neuroprotection. The secosteroid 1alpha,25-dihydroxyvitamin D3 (1,25-D3) is a modulator of the immune system and also exhibits a strong potential for neuroprotection as recently shown in the MCAO model of cerebral ischemia. We studied the effects of 1,25-D3 treatment on heme oxygenase-1 expression following focal cortical ischemia elicited by photothrombosis. Postlesional treatment with 1,25-D3 (4 microg/kg body weight) resulted in a transient, but significant upregulation of glial heme oxygenase-1 immunoreactivity concomitant with a reduction in glial fibrillary acidic protein immunoreactivity in remote cortical regions affected by a secondary spread of injury, whereas the size of the lesion's core remained unaffected. 1,25-D3 did not produce a temporal shift or extension of injury-related heme oxygenase-1 responses, indicating that 1,25-D3 did not prolong ischemia-related heme oxygenase-1 expression. In contrast to glial heme oxygenase-1 upregulation, glial fibrillary acidic protein, a sensitive marker for reactive gliosis, was significantly reduced. These findings support an additional protective action of 1,25-D3 at the cellular level in regions affected by secondary injury-related responses.


Subject(s)
Calcitriol/pharmacology , Cerebral Cortex/metabolism , Glial Fibrillary Acidic Protein/genetics , Heme Oxygenase (Decyclizing)/genetics , Intracranial Thrombosis/metabolism , Animals , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Fluorescent Dyes , Heme Oxygenase-1 , Intracranial Thrombosis/pathology , Male , Rats , Rats, Wistar , Rose Bengal
SELECTION OF CITATIONS
SEARCH DETAIL
...