Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Int J Mol Sci ; 24(23)2023 Nov 21.
Article En | MEDLINE | ID: mdl-38068900

S100B, a homodimeric Ca2+-binding protein, is produced and secreted by astrocytes, and its extracellular levels have been used as a glial marker in brain damage and neurodegenerative and psychiatric diseases; however, its mechanism of secretion is elusive. We used primary astrocyte cultures and calcium measurements from real-time fluorescence microscopy to investigate the role of intracellular calcium in S100B secretion. In addition, the dimethyl sulfoxide (DMSO) effect on S100B was investigated in vitro and in vivo using Wistar rats. We found that DMSO, a widely used vehicle in biological assays, is a powerful S100B secretagogue, which caused a biphasic response of Ca2+ mobilization. Our data show that astroglial S100B secretion is triggered by the increase in intracellular Ca2+ and indicate that this increase is due to Ca2+ mobilization from the endoplasmic reticulum. Also, blocking plasma membrane Ca2+ channels involved in the Ca2+ replenishment of internal stores decreased S100B secretion. The DMSO-induced S100B secretion was confirmed in vivo and in ex vivo hippocampal slices. Our data support a nonclassic vesicular export of S100B modulated by Ca2+, and the results might contribute to understanding the mechanism underlying the astroglial release of S100B.


Astrocytes , Dimethyl Sulfoxide , Rats , Animals , Rats, Wistar , Dimethyl Sulfoxide/pharmacology , Dimethyl Sulfoxide/metabolism , Astrocytes/metabolism , Colforsin/pharmacology , Secretagogues/pharmacology , Calcium/metabolism , Nerve Growth Factors/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Endoplasmic Reticulum/metabolism , Cells, Cultured
2.
J Alzheimers Dis ; 84(4): 1415-1430, 2021.
Article En | MEDLINE | ID: mdl-34719501

One of the changes found in the brain in Alzheimer's disease (AD) is increased calpain, derived from calcium dysregulation, oxidative stress, and/or neuroinflammation, which are all assumed to be basic pillars in neurodegenerative diseases. The role of calpain in synaptic plasticity, neuronal death, and AD has been discussed in some reviews. However, astrocytic calpain changes sometimes appear to be secondary and consequent to neuronal damage in AD. Herein, we explore the possibility of calpain-mediated astroglial reactivity in AD, both preceding and during the amyloid phase. We discuss the types of brain calpains but focus the review on calpains 1 and 2 and some important targets in astrocytes. We address the signaling involved in controlling calpain expression, mainly involving p38/mitogen-activated protein kinase and calcineurin, as well as how calpain regulates the expression of proteins involved in astroglial reactivity through calcineurin and cyclin-dependent kinase 5. Throughout the text, we have tried to provide evidence of the connection between the alterations caused by calpain and the metabolic changes associated with AD. In addition, we discuss the possibility that calpain mediates amyloid-ß clearance in astrocytes, as opposed to amyloid-ß accumulation in neurons.


Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Astrocytes/metabolism , Brain/metabolism , Calpain/metabolism , Neuronal Plasticity , Alzheimer Disease/pathology , Animals , Astrocytes/pathology , Calcineurin/metabolism , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Disease Models, Animal , Humans , Neuroinflammatory Diseases/metabolism
3.
Brain Res ; 1698: 54-61, 2018 11 01.
Article En | MEDLINE | ID: mdl-29932894

Sepsis is one of the main causes of hospitalization and mortality in Intensive Care Units. One of the first manifestations of sepsis is encephalopathy, reported in up to 70% of patients, being associated with higher mortality and morbidity. The factors that cause sepsis-associated encephalopathy (SAE) are still not well known, and may be multifactorial, as perfusion changes, neuroinflammation, oxidative stress and glycolytic metabolism alterations. Fructose-1,6-bisphosphate (FBP), a metabolite of the glycolytic route, has been reported as neuroprotective agent. The present study used an experimental sepsis model in C57BL/6 mice. We used in vivo brain imaging to evaluate glycolytic metabolism through microPET scans and the radiopharmaceutical 18F-fluoro-2-deoxy-D-glucose (18F-FDG). Brain images were obtained before and 12 h after the induction of sepsis in animals with and without FBP treatment. We also evaluated the treatment effects in the brain oxidative stress by measuring the production of reactive oxygen species (ROS), the activity of catalase (CAT) and glutathione peroxidase (GPx), and the levels of fluorescent marker 2'7'-dichlorofluorescein diacetate (DCF). There was a significant decrease in brain glucose metabolism due to experimental sepsis. A significant protective effect of FBP treatment was observed in the cerebral metabolic outcomes. FBP also modulated the production of ROS, evidenced by reduced CAT activity and lower levels of DCF. Our results suggest that FBP may be a possible candidate in the treatment of SAE.


Fructosediphosphates/pharmacology , Glucose/metabolism , Reactive Oxygen Species/metabolism , Sepsis/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain Diseases/drug therapy , Disease Models, Animal , Fluorodeoxyglucose F18 , Fructose/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Positron Emission Tomography Computed Tomography/methods , Sepsis/drug therapy
4.
Front Neurosci ; 12: 1035, 2018.
Article En | MEDLINE | ID: mdl-30728759

Based on the concept of the tripartite synapse, we have reviewed the role of glucose-derived compounds in glycolytic pathways in astroglial cells. Glucose provides energy and substrate replenishment for brain activity, such as glutamate and lipid synthesis. In addition, glucose metabolism in the astroglial cytoplasm results in products such as lactate, methylglyoxal, and glutathione, which modulate receptors and channels in neurons. Glucose has four potential destinations in neural cells, and it is possible to propose a crossroads in "X" that can be used to describe these four destinations. Glucose-6P can be used either for glycogen synthesis or the pentose phosphate pathway on the left and right arms of the X, respectively. Fructose-6P continues through the glycolysis pathway until pyruvate is formed but can also act as the initial compound in the hexosamine pathway, representing the left and right legs of the X, respectively. We describe each glucose destination and its regulation, indicating the products of these pathways and how they can affect synaptic communication. Extracellular L-lactate, either generated from glucose or from glycogen, binds to HCAR1, a specific receptor that is abundantly localized in perivascular and post-synaptic membranes and regulates synaptic plasticity. Methylglyoxal, a product of a deviation of glycolysis, and its derivative D-lactate are also released by astrocytes and bind to GABAA receptors and HCAR1, respectively. Glutathione, in addition to its antioxidant role, also binds to ionotropic glutamate receptors in the synaptic cleft. Finally, we examined the hexosamine pathway and evaluated the effect of GlcNAc-modification on key proteins that regulate the other glucose destinations.

5.
Mol Neurobiol ; 54(8): 6356-6377, 2017 Oct.
Article En | MEDLINE | ID: mdl-27722926

The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induces motor and nonmotor dysfunctions resembling Parkinson's disease (PD); however, studies investigating the effects of 1-methyl-4-phenylpyridinium (MPP+), an active oxidative product of MPTP, are scarce. This study investigated the behavioral and striatal neurochemical changes (related to oxidative damage, glial markers, and neurotrophic factors) 24 h after intracerebroventricular administration of MPP+ (1.8-18 µg/mouse) in C57BL6 mice. MPP+ administration at high dose (18 µg/mouse) altered motor parameters, since it increased the latency to leave the first quadrant and reduced crossing, rearing, and grooming responses in the open-field test and decreased rotarod latency time. MPP+ administration at low dose (1.8 µg/mouse) caused specific nonmotor dysfunctions as it produced a depressive-like effect in the forced swim test and tail suspension test, loss of motivational and self-care behavior in the splash test, anxiety-like effect in the elevated plus maze test, and short-term memory deficit in the step-down inhibitory avoidance task, without altering ambulation. MPP+ at doses of 1.8-18 µg/mouse increased tyrosine hydroxylase (TH) immunocontent and at 18 µg/mouse increased α-synuclein and decreased parkin immunocontent. The astrocytic calcium-binding protein S100B and glial fibrillary acidic protein (GFAP)/S100B ratio was decreased following MPP+ administration (18 µg/mouse). At this highest dose, MPP+ increased the ionized calcium-binding adapter molecule 1 (Iba-1) immunocontent, suggesting microglial activation. Also, MPP+ at a dose of 18 µg/mouse increased thiobarbituric acid reactive substances (TBARS) and glutathione (GSH) levels and increased glutathione peroxidase (GPx) and hemeoxygenase-1 (HO-1) immunocontent, suggesting a significant role for oxidative stress in the MPP+-induced striatal damage. MPP+ (18 µg/mouse) also increased striatal fibroblast growth factor 2 (FGF-2) and brain-derived neurotrophic factor (BDNF) levels. Moreover, MPP+ decreased tropomyosin receptor kinase B (TrkB) immunocontent. Finally, MPP+ (1.8-18 µg/mouse) increased serum corticosterone levels and did not alter acetylcholinesterase (AChE) activity in the striatum but increased it in cerebral cortex and hippocampus. Collectively, these results indicate that MPP+ administration at low doses may be used as a model of emotional and memory/learning behavioral deficit related to PD and that MPP+ administration at high dose could be useful for analysis of striatal dysfunctions associated with motor deficits in PD.


1-Methyl-4-phenylpyridinium/toxicity , Corpus Striatum/drug effects , Emotions/drug effects , Learning/drug effects , Memory/drug effects , Motor Activity/drug effects , Acetylcholinesterase/metabolism , Animals , Corpus Striatum/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glutathione/metabolism , Mice , S100 Calcium Binding Protein beta Subunit/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
6.
PLoS One ; 10(5): e0127845, 2015.
Article En | MEDLINE | ID: mdl-25978644

The developing brain is vulnerable to the effects of ethanol. Glutamate is the main mediator of excitatory signals in the brain and is probably involved in most aspects of normal brain function during development. The aim of this study was to investigate vulnerability to and the impact of ethanol toxicity on glutamate uptake signaling in adolescent rats after moderate pre and postnatal ethanol exposure. Pregnant female rats were divided into three groups and treated only with water (control), non-alcoholic beer (vehicle) or 10% (v/v) beer solution (moderate prenatal alcohol exposure-MPAE). Thirty days after birth, adolescent male offspring were submitted to hippocampal acute slice procedure. We assayed glutamate uptake and measured glutathione content and also quantified glial glutamate transporters (EAAT 1 and EAAT 2). The glutamate system vulnerability was tested with different acute ethanol doses in naïve rats and compared with the MPAE group. We also performed a (lipopolysaccharide-challenge (LPS-challenge) with all groups to test the glutamate uptake response after an insult. The MPAE group presented a decrease in glutamate uptake corroborating a decrease in glutathione (GSH) content. The reduction in GSH content suggests oxidative damage after acute ethanol exposure. The glial glutamate transporters were also altered after prenatal ethanol treatment, suggesting a disturbance in glutamate signaling. This study indicates that impairment of glutamate uptake can be dose-dependent and the glutamate system has a higher vulnerability to ethanol toxicity after moderate ethanol exposure In utero. The effects of pre- and postnatal ethanol exposure can have long-lasting impacts on the glutamate system in adolescence and potentially into adulthood.


Ethanol/adverse effects , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Animals , Female , Glutathione/metabolism , Male , Pregnancy , Rats
7.
Psychiatry Res ; 220(3): 745-51, 2014 Dec 30.
Article En | MEDLINE | ID: mdl-25457283

The role of cholesterol in psychiatric diseases has aroused the interest of the medical community, particularly in association with violent and suicidal behavior. Herein, we discuss some aspects of brain cholesterol metabolism, exploring possible mechanisms underlying the findings and reviewing the available literature on the possible neurochemical link between suicide and low or reduced levels of serum cholesterol. Most of the current hypotheses suggest a decreased serotonergic activity due to a decrease in cholesterol in the lipid rafts of synaptic membranes. Some aspects and limitations of this assumption are emphasized. In addition to serotonin hypofunction, other mechanisms have been proposed to explain increased impulsivity in suicidal individuals, including steroid modulation and brain-derived neurotrophic factor decrease, which could also be related to changes in lipid rafts. Other putative markers of suicidal behavior (e.g. protein S100B) are discussed in connection with cholesterol metabolism in the brain tissue.


Brain/metabolism , Cholesterol/metabolism , Mental Disorders/metabolism , Suicidal Ideation , Suicide , Biomarkers/metabolism , Brain-Derived Neurotrophic Factor/blood , Cholesterol/blood , Humans , Mental Disorders/diagnosis , Mental Disorders/psychology , Serotonin/blood , Serotonin/metabolism , Suicide/psychology
8.
J Neuroimmunol ; 249(1-2): 8-15, 2012 Aug 15.
Article En | MEDLINE | ID: mdl-22560157

In this study we investigated the anti-inflammatory effects of chronic ethanol (EtOH) treatment on lipopolysaccharide (LPS)-stimulated C6 glioma cells. The cells were chronically treated with 200mM EtOH; coincubation with LPS and EtOH was obtained upon addition of 2µg/ml LPS to the incubation medium in the last 24h of EtOH exposure. We found that EtOH prevented the LPS-induced production of tumor necrosis factor α (TNFα) without decreasing cell viability. Either LPS treated or EtOH plus LPS treated cells presented upregulated glial fibrillary acidic protein (GFAP) and downregulated vimentin levels characterizing a program of reactive astrogliosis. Also, EtOH plus LPS stimulation greatly increased the oxidative stress generation evaluated by DCF-DA measurement, while either EtOH alone or LPS alone was unable to induce oxidative stress. Western blot analysis indicated that either EtOH, LPS or EtOH plus LPS treatments are unable to affect Akt/GSK3ß signaling pathway. However, LPS alone and EtOH plus LPS co-treatment inhibited Erk phosphorylation. A dramatic loss of stress fibers was found in EtOH exposed cells, evaluated by cytochemistry using phalloidin-fluorescein. However, LPS alone was not able to disrupt actin organization. Furthermore, cells co-incubated with LPS and EtOH presented reversion of the disrupted stress fibers provoked by EtOH. Supporting this action, RhoA and vinculin immunocontent were upregulated in response to EtOH plus LPS. Interestingly, EtOH suppresses the inflammatory cascade (TNFα production) in response to LPS. Concomitantly it sustains Erk inhibition, increases oxidative stress generation and induces reactive astrogliosis in the presence of LPS, conditions associated with neurotoxicity. The effects observed were not supported by actin reorganization. Altogether, these findings suggest that Erk signaling inhibition could play a role in both suppressing TNFα production and inducing oxidative stress generation and astrogliosis, therefore modulating a dual action of EtOH plus LPS in glial cells.


Anti-Inflammatory Agents/pharmacology , Ethanol/pharmacology , MAP Kinase Signaling System/drug effects , Neuroglia/drug effects , Oxidative Stress/drug effects , Animals , Blotting, Western , Cell Line, Tumor , Cell Survival , Electrophoresis, Polyacrylamide Gel , Gliosis/chemically induced , Gliosis/metabolism , Gliosis/pathology , Immunohistochemistry , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/toxicity , Neuroglia/metabolism , Neuroglia/pathology , Rats , Stress Fibers/drug effects , Stress Fibers/metabolism , Tumor Necrosis Factor-alpha/metabolism
9.
J Neuroinflammation ; 8: 128, 2011 Oct 04.
Article En | MEDLINE | ID: mdl-21970823

BACKGROUND: Inflammatory responses in brain are primarily mediated by microglia, but growing evidence suggests a crucial importance of astrocytes. S100B, a calcium-binding protein secreted by astrocytes, has properties of a neurotrophic or an inflammatory cytokine. However, it is not known whether primary signals occurring during induction of an inflammatory response (e.g. lipopolysaccharide, LPS) directly modulate S100B. METHODS: In this work, we evaluated whether S100B levels in cerebrospinal fluid (CSF) and serum of Wistar rats are affected by LPS administered by intraperitoneal (IP) or intracerebroventricular (ICV) injection, as well as whether primary astrocyte cultures respond directly to lipopolysaccharide. RESULTS: Our data suggest that S100B secretion in brain tissue is stimulated rapidly and persistently (for at least 24 h) by ICV LPS administration. This increase in CSF S100B was transient when LPS was IP administered. In contrast to these S100B results, we observed an increase in in TNFα levels in serum, but not in CSF, after IP administration of LPS. In isolated astrocytes and in acute hippocampal slices, we observed a direct stimulation of S100B secretion by LPS at a concentration of 10 µg/mL. An involvement of TLR4 was confirmed by use of specific inhibitors. However, lower levels of LPS in astrocyte cultures were able to induce a decrease in S100B secretion after 24 h, without significant change in intracellular content of S100B. In addition, after 24 h exposure to LPS, we observed a decrease in astrocytic glutathione and an increase in astrocytic glial fibrillary acidic protein. CONCLUSIONS: Together, these data contribute to the understanding of the effects of LPS on astrocytes, particularly on S100B secretion, and help us to interpret cerebrospinal fluid and serum changes for this protein in neuroinflammatory diseases. Moreover, non-brain S100B-expressing tissues may be differentially regulated, since LPS administration did not lead to increased serum levels of S100B.


Astrocytes/drug effects , Astrocytes/metabolism , Lipopolysaccharides/pharmacology , Nerve Growth Factors/cerebrospinal fluid , Nerve Growth Factors/metabolism , S100 Proteins/cerebrospinal fluid , S100 Proteins/metabolism , Animals , Astrocytes/cytology , Cells, Cultured , Cerebral Cortex/cytology , Glutathione/metabolism , Infusions, Intraventricular , Lipopolysaccharides/administration & dosage , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/cerebrospinal fluid
10.
J Neural Transm (Vienna) ; 117(11): 1295-305, 2010 Nov.
Article En | MEDLINE | ID: mdl-20953641

The intracerebroventricular infusion of streptozotocin (icv-STZ) has been largely used in research to mimic the main characteristics of Alzheimer's disease (AD), including cognitive decline, impairment of cholinergic transmission, oxidative stress and astrogliosis. Moderate physical exercise has a number of beneficial effects on the central nervous system, as demonstrated both in animals and in human studies. This study aimed to evaluate the effect of 5-week treadmill training, in the icv-SZT model of sporadic AD, on cognitive function, oxidative stress (particularly mediated by NO) and on the astrocyte marker proteins, glial fibrillary acidic protein (GFAP) and S100B. Results confirm the spatial cognitive deficit and oxidative stress in this model, as well as astroglial alterations, particularly a decrease in CSF S100B. Physical exercise prevented these alterations, as well as increasing the hippocampal content of glutathione and GFAP per se in the CA1 region. These findings reinforce the potential neuroprotective role of moderate physical exercise. Astroglial changes observed in this dementia model contribute to understanding AD and other diseases that are accompanied by cognitive deficit.


Cognition Disorders/rehabilitation , Hippocampus/physiology , Motor Activity/physiology , Oxidative Stress/physiology , Alzheimer Disease/rehabilitation , Animals , Cognition Disorders/chemically induced , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Glutathione/metabolism , Hippocampus/drug effects , Immunohistochemistry , Injections, Intraventricular , Male , Maze Learning/physiology , Neurotoxins/administration & dosage , Neurotoxins/toxicity , Rats , Rats, Wistar , Streptozocin/administration & dosage , Streptozocin/toxicity
11.
Int J Dev Neurosci ; 28(2): 153-9, 2010 Apr.
Article En | MEDLINE | ID: mdl-20036324

We have previously demonstrated that early environment influences the metabolic response, affecting abdominal fat deposition in adult female rats exposed to a long-term highly caloric diet. In the present study, our goal was to verify the effects of the chronic exposure, in adulthood, to a highly palatable diet (chocolate) on cerebral Na+,K+-ATPase activity and S100B protein concentrations, and the response to its withdrawal in neonatally handled and non-handled rats. We measured the consumption of foods (standard lab chow and chocolate), body weight gain, S100B protein concentrations, as well as cerebral Na(+),K(+)-ATPase activity during chronic exposure and after chocolate withdrawal in adult female rats that had been exposed or not to neonatal handling (10 min/day, 10 first days of life). Non-handled rats chronically exposed to chocolate exhibited increased plasma S100B levels, but there was no difference in abdominal fat S100B concentration between groups. Chronic chocolate consumption decreased Na+,K+-ATPase activity in both amygdala and hippocampus in non-handled, but not in handled rats, and this effect disappeared after chocolate withdrawal. Non-handled animals also demonstrated increased frequency of head shaking in the open field after 24h of chocolate withdrawal in comparison to handled ones. These findings suggest that neonatal handling modifies the vulnerability to metabolic and brain alterations induced by chronic exposure to a highly palatable diet in adulthood.


Aging/metabolism , Brain/enzymology , Dietary Carbohydrates/metabolism , Food Deprivation/physiology , Nerve Growth Factors/metabolism , S100 Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Animals, Newborn , Female , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit
12.
J Alzheimers Dis ; 17(1): 193-202, 2009.
Article En | MEDLINE | ID: mdl-19494442

Although the exact cause of Alzheimer's disease remains elusive, many possible risk factors and pathological alterations have been used in the elaboration of in vitro and in vivo models of this disease in rodents, including intracerebral infusion of streptozotocin (STZ). Using this model, we evaluated spatial cognitive deficit and neurochemical hippocampal alterations, particularly astroglial protein markers such as glial fibrillary acidic protein (GFAP) and S100B, glutathione content, nitric oxide production, and cerebrospinal fluid (CSF) S100B. In addition, prevention of these alterations by aminoguanidine administration was evaluated. Results confirm a spatial cognitive deficit and nitrative stress in this dementia model as well as specific astroglial alterations, particularly S100B accumulation in the hippocampus and decreased CSF S100B. The hippocampal astroglial activation occurred independently of the significant alteration in GFAP content. Moreover, all these alterations were completely prevented by aminoguanidine administration, confirming the neuroprotective potential of this compound, but suggesting that nitrative stress and/or glycation may be underlying these alterations. These findings contribute to the understanding of diseases accompanied by cognitive deficits and the STZ-model of dementia.


Dementia , Enzyme Inhibitors/therapeutic use , Guanidines/therapeutic use , Hippocampus/pathology , Streptozocin , Analysis of Variance , Animals , Behavior, Animal/drug effects , Dementia/chemically induced , Dementia/pathology , Dementia/prevention & control , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Glutathione/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Maze Learning/drug effects , Nerve Growth Factors/cerebrospinal fluid , Nitric Oxide/metabolism , Rats , Rats, Wistar , Reaction Time/drug effects , S100 Calcium Binding Protein beta Subunit , S100 Proteins/cerebrospinal fluid
13.
Neurochem Res ; 34(9): 1603-11, 2009 Sep.
Article En | MEDLINE | ID: mdl-19288274

Hippocampal slices have been widely used to investigate electrophysiological and metabolic neuronal parameters, as well as parameters of astroglial activity including protein phosphorylation and glutamate uptake. S100B is an astroglial-derived protein, which extracellularly plays a neurotrophic activity during development and excitotoxic insult. Herein, we characterized S100B secretion in acute hippocampal slices exposed to different concentrations of K(+) and Ca(2+) in the extracellular medium. Absence of Ca(2+) and/or low K(+) (0.2 mM KCl) caused an increase in S100B secretion, possibly by mobilization of internal stores of Ca(2+). In contrast, high K(+) (30 mM KCl) or calcium channel blockers caused a decrease in S100B secretion. This study suggests that exposure of acute hippocampal slices to low- and high-K(+) could be used as an assay to evaluate astrocyte activity by S100B secretion: positively regulated by low K(+) (possibly involving mobilization of internal stores of Ca(2+)) and negatively regulated by high-K(+) (likely secondary to influx of K(+)).


Calcium/metabolism , Hippocampus/metabolism , Nerve Growth Factors/metabolism , Potassium/metabolism , S100 Proteins/metabolism , Animals , Calcium/pharmacology , Calcium Channel Blockers/pharmacology , Extracellular Space/metabolism , Glutamic Acid/metabolism , Glutathione/metabolism , Hippocampus/drug effects , Male , Phosphopyruvate Hydratase/metabolism , Potassium/pharmacology , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit , Verapamil/pharmacology
14.
Brain Res ; 1251: 204-12, 2009 Jan 28.
Article En | MEDLINE | ID: mdl-19056357

The permanent occlusion of common carotid arteries (2VO) causes a significant reduction of cerebral blood flow (hypoperfusion) in rats and constitutes a well established experimental model to investigate neuronal damage and cognitive impairment that occurs in human ageing and Alzheimer's disease. In the present study, we evaluated two astroglial proteins--S100B and glial fibrillary acidic protein (GFAP)--in cerebral cortex and hippocampus tissue, glutamate uptake and glutamine synthetase activity in hippocampus tissue, as well as S100B in cerebrospinal fluid. Cognition, as assessed by reference and working spatial memory protocols, was also investigated. Adult male Wistar rats were submitted to 10 weeks of chronic cerebral hypoperfusion by the 2VO method. A significant increase of S100B and GFAP in hippocampus tissue was observed, as well a significant decrease in glutamate uptake. Interestingly, we observed a decrease in S100B in cerebrospinal fluid. As for the cognitive outcome, there was an impairment of both reference and working spatial memory in the water maze; positive correlation between cognitive impairment and glutamate uptake decrease was evidenced in hypoperfused rats. These data support the hypothesis that astrocytes play a crucial role in the mechanisms of experimental neurodegeneration and that hippocampal pathology arising after chronic hypoperfusion gives rise to memory deficits.


Astrocytes/metabolism , Cerebral Cortex/physiopathology , Cognition Disorders/physiopathology , Gliosis/physiopathology , Hypoxia-Ischemia, Brain/physiopathology , Hypoxia-Ischemia, Brain/psychology , Animals , Astrocytes/pathology , Biomarkers/analysis , Biomarkers/metabolism , Carotid Stenosis/metabolism , Carotid Stenosis/physiopathology , Carotid Stenosis/psychology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cognition Disorders/etiology , Cognition Disorders/metabolism , Disease Models, Animal , Glial Fibrillary Acidic Protein/analysis , Glial Fibrillary Acidic Protein/metabolism , Gliosis/etiology , Gliosis/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamic Acid/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Hypoxia-Ischemia, Brain/metabolism , Male , Maze Learning/physiology , Memory Disorders/etiology , Memory Disorders/metabolism , Memory Disorders/physiopathology , Nerve Growth Factors/analysis , Nerve Growth Factors/metabolism , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit , S100 Proteins/analysis , S100 Proteins/metabolism , Synaptic Transmission/physiology
15.
J Neuroimmunol ; 206(1-2): 52-7, 2009 Jan 03.
Article En | MEDLINE | ID: mdl-19042033

S100B is an astrocyte-derived cytokine implicated in the IL-1beta-triggered cytokine cycle in Alzheimer's disease. However, the secretion of S100B following stimulation by IL-1beta has not been directly demonstrated. We investigated S100B secretion in cortical primary astrocyte cultures, C6 glioma cells and acute hippocampal slices exposed to IL-1beta. S100B secretion was induced by IL-1beta in all preparations, involving MAPK pathway and, apparently, NF-small ka, CyrillicB signaling. Astrocytes and C6 cells exhibited different sensitivities to IL-1beta. These results suggest that IL-1beta-induced S100B secretion is a component of the neuroinflammatory response, which would support the involvement of S100B in the genesis of neurodegenerative diseases.


Hippocampus/drug effects , Interleukin-1beta/pharmacology , Mitogen-Activated Protein Kinase Kinases/metabolism , Nerve Growth Factors/metabolism , Neuroglia/drug effects , S100 Proteins/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Indoles , L-Lactate Dehydrogenase/metabolism , Nitric Oxide/metabolism , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit , Signal Transduction/drug effects , Subcellular Fractions/drug effects , Time Factors
16.
Prog Neuropsychopharmacol Biol Psychiatry ; 32(6): 1580-3, 2008 Aug 01.
Article En | MEDLINE | ID: mdl-18582527

S100B is a calcium-binding protein, produced and secreted by astrocytes, which has a putative paracrine neurotrophic activity. Clinical studies have suggested that peripheral elevation of this protein is positively correlated with a therapeutic antidepressant response, particularly to selective serotonin reuptake inhibitors (SSRIs); however, the mechanism underlying this response remains unclear. Here, we measured S100B secretion directly in hippocampal astrocyte cultures and hippocampal slices exposed to fluoxetine and observed a significant increment of S100B release in the presence of this SSRI, apparently dependent on protein kinase A (PKA). Moreover, we found that serotonin (possibly via the 5HT1A receptor) reduces S100B secretion and antagonizes the effect of fluoxetine on S100B secretion. These data reinforce the effect of fluoxetine, independently of serotonin and serotonin receptors, suggesting a putative role for S100B in depressive disorders and suggesting that other molecular targets may be relevant for antidepressant activity.


Astrocytes/metabolism , Fluoxetine/pharmacology , Nerve Growth Factors/metabolism , S100 Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin/physiology , Animals , Astrocytes/drug effects , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Rats , Rats, Wistar , Receptors, Serotonin/drug effects , S100 Calcium Binding Protein beta Subunit , Serotonin/pharmacology
17.
Behav Brain Res ; 190(2): 206-11, 2008 Jul 19.
Article En | MEDLINE | ID: mdl-18395808

Lesion of the nucleus basalis magnocellularis (nbm) is a suitable approach to study cognitive deficit and behavior alterations involving cholinergic dysfunction, which is associated with the major types of dementia. Cortical astrogliosis also has been described in this model, but it is not clear whether hippocampal astrocytes are activated. In this study, we investigated possible specific astrocyte alterations in the hippocampi of Wistar rats submitted to nbm damage with ibotenic acid, investigating the content and immunohistochemistry of glial fibrillary acidic protein (GFAP), as well as S100B protein content, glutamate uptake and glutamine synthetase activity on the 7th and 28th post-lesion days. Cognitive deficit was confirmed by the step-down inhibitory avoidance task. Interestingly, we found a decrease in GFAP content, S100B content and glutamate uptake activity in the hippocampus on the 28th day after nbm lesion. No alterations were observed in glutamine synthetase activity or in the cerebrospinal fluid S100B content. Although our data suggest caution in the use of nbm lesion with ibotenic acid as a dementia model, it is possible that these alterations could contribute to the cognitive deficit observed in these rats.


Astrocytes/cytology , Avoidance Learning/physiology , Basal Nucleus of Meynert/physiology , Cholinergic Fibers/metabolism , Dementia/physiopathology , Hippocampus/cytology , Animals , Astrocytes/metabolism , Basal Nucleus of Meynert/cytology , Basal Nucleus of Meynert/drug effects , Brain Damage, Chronic/chemically induced , Cell Count , Dementia/metabolism , Disease Models, Animal , Exploratory Behavior/physiology , Follow-Up Studies , Glial Fibrillary Acidic Protein/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamic Acid/metabolism , Habituation, Psychophysiologic/physiology , Hippocampus/metabolism , Ibotenic Acid , Immunohistochemistry , Male , Nerve Growth Factors/metabolism , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Statistics, Nonparametric , Time Factors
18.
Brain Res ; 1187: 33-41, 2008 Jan 02.
Article En | MEDLINE | ID: mdl-18021757

Pre- and postnatal protein malnutrition (PMN) adversely affects the developing brain in numerous ways, but only a few studies have investigated specific glial parameters. This study aimed to evaluate specific glial changes of rats exposed to pre and postnatal PMN, based on glial fibrillary acidic protein (GFAP) and S100B immunocontents as well as glutamine synthetase (GS), in cerebral cortex, hippocampus, cerebellum and cerebrospinal fluid, on the 2nd, 15th and 60th postnatal days. We found increases in GFAP, S100B and GS in the cerebral cortex at birth, suggesting an astrogliosis. Hippocampus and cerebellum also exhibited this profile at birth. However, a significant interaction between age and diet in postnatal life was observed only in the S100B of the cerebral cortex. No changes in the content of GFAP and S100B and GS activity were found on the 60th postnatal day in malnourished rats. In contrast, following an increase in the levels of S100B in the cerebrospinal fluid, during the early developmental stages, levels remained elevated on the 60th postnatal day. Our data support the concept of astrogliosis at birth, induced by PMN, and involve extracellular-regulated kinase activation. Specific alterations in cerebral cortex emphasize the regional vulnerability of the brain to malnutrition; some alterations were observed only at birth (e.g. GFAP); others were observed on the 2nd and 15th post-natal days (e.g. ERK phosphorylation). Taken together, transient and persistent alterations (e.g. elevated extracellular levels of S100B) suggest some brain damage or a risk of brain diseases in rats exposed to PMN.


Brain Damage, Chronic/etiology , Brain Damage, Chronic/physiopathology , Fetal Nutrition Disorders/physiopathology , Gliosis/etiology , Gliosis/physiopathology , Protein Deficiency/physiopathology , Aging/metabolism , Animals , Animals, Newborn , Biomarkers/analysis , Biomarkers/cerebrospinal fluid , Brain/embryology , Brain/growth & development , Brain/physiopathology , Brain Damage, Chronic/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glial Fibrillary Acidic Protein/cerebrospinal fluid , Gliosis/metabolism , Glutamate-Ammonia Ligase/cerebrospinal fluid , Male , Nerve Growth Factors/cerebrospinal fluid , Neuroglia/metabolism , Pregnancy , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit , S100 Proteins/cerebrospinal fluid
19.
Clin Exp Pharmacol Physiol ; 34(11): 1126-30, 2007 Nov.
Article En | MEDLINE | ID: mdl-17880365

1. S100B is a calcium-binding protein that acts as a neurotrophic cytokine and is expressed in the central nervous system, predominantly by astrocytes. At nanomolar concentrations, S100B stimulates neurite outgrowth and glial glutamate uptake, as well as protecting neurons against glutamate excitoxicity. 2. Peripheral S100B concentrations, particularly in the serum and cerebrospinal fluid (CSF), have been used as a parameter of glial activation or death in several physiological and pathological conditions. 3. In the present study, we investigated the effect of anaesthetics (thiopental, ketamine and halothane) on CSF concentrations of S100B, as well as a possible sex dependence, because several studies have suggested astrocytes as putative targets for oestrogen. 4. Higher levels of CSF S100B were found when rats were anaesthetized with thiopental; these levels, independently of anaesthetic, were sex dependent. Conversely, no effect of anaesthetic or sex was observed on serum concentrations of S100B. 5. The increase in CSF concentrations of S100B induced by thiopental was confirmed in non-anaesthetized neonatal rats and cortical astrocyte cultures. 6. Assuming CSF S100B as a marker of development, glial activation or even brain damage, investigations regarding the sex dependence of its concentration may be useful in gaining an understanding of sex variations in the behaviour and the pathological course of, as well as susceptibility to, many brain disorders. The findings of the present study reinforce the sex effect on synaptic plasticity and suggest a sex dependence of neural communication mediated by extracellular S100B without restricting the influence of astrocytes on the developmental phase.


Anesthetics/pharmacology , Astrocytes/drug effects , Cerebral Cortex/drug effects , Cisterna Magna/drug effects , Halothane/pharmacology , Ketamine/pharmacology , Nerve Growth Factors/cerebrospinal fluid , S100 Proteins/cerebrospinal fluid , Thiopental/pharmacology , Animals , Animals, Newborn , Astrocytes/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cisterna Magna/metabolism , Female , Male , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit , Sex Factors
20.
Brain Res ; 1144: 107-16, 2007 May 04.
Article En | MEDLINE | ID: mdl-17335785

This study was undertaken to verify if repeated long-term separation from dams would affect the development of parameters related to post-traumatic stress disorder (PTSD) after animals are subjected to inescapable shock when adults. Wistar rats were subjected to repeated maternal separation during post-natal days 1-10. When adults, rats from both sexes were submitted to a PTSD model consisting of exposure to inescapable footshock, followed by situational reminders. We observed long-lasting effects of both interventions. Exposure to shock increased fear conditioning. Anxiety-like behavior was increased and exploratory activity decreased by both treatments, and these effects were more robust in males. Additionally, basal corticosterone in plasma was decreased, paralleling effects observed in PTSD patients. Levels of S100B protein in serum and cerebrospinal fluid (CSF) were measured. Levels in serum correlated with the effects observed in anxiety-like behavior, increasing in males exposed to shock, and presenting no effect in females. S100B in CSF was increased in females submitted to maternal separation during the neonatal period. These results suggest that, in rats, an early stress experience such as maternal separation may aggravate some effects of exposure to a stressor during adult age, and that this effect is sex-specific. Additionally, data suggest that the increased S100B levels, observed in serum, have an extracerebral origin, possibly mediated by an increase in the noradrenergic tonus. Increased S100B in brain could be related to its neurotrophic actions.


Behavior, Animal/physiology , Maternal Deprivation , Nerve Growth Factors/metabolism , S100 Proteins/metabolism , Sex Characteristics , Stress Disorders, Post-Traumatic/metabolism , Analysis of Variance , Animals , Animals, Newborn , Conditioning, Psychological/physiology , Corticosterone/blood , Disease Models, Animal , Electroshock/adverse effects , Exploratory Behavior/physiology , Fear/physiology , Female , Male , Pregnancy , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit
...