Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Biochimie ; 219: 21-32, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37541567

ABSTRACT

Non ketotic hyperglycinemia (NKH) is an inborn error of glycine metabolism caused by mutations in the genes encoding glycine cleavage system proteins. Classic NKH has a neonatal onset, and patients present with severe neurodegeneration. Although glycine accumulation has been implicated in NKH pathophysiology, the exact mechanisms underlying the neurological damage and white matter alterations remain unclear. We investigated the effects of glycine in the brain of neonatal rats and MO3.13 oligodendroglial cells. Glycine decreased myelin basic protein (MBP) and myelin-associated glycoprotein (MAG) in the corpus callosum and striatum of rats on post-natal day (PND) 15. Glycine also reduced neuroglycan 2 (NG2) and N-methyl-d-aspartate receptor subunit 1 (NR1) in the cerebral cortex and striatum on PND15. Moreover, glycine reduced striatal glutamate aspartate transporter 1 (GLAST) content and neuronal nucleus (NeuN), and increased glial fibrillary acidic protein (GFAP) on PND15. Glycine also increased DCFH oxidation and malondialdehyde levels and decreased GSH concentrations in the cerebral cortex and striatum on PND6, but not on PND15. Glycine further reduced viability but did not alter DCFH oxidation and GSH levels in MO3.13 cells after 48- and 72-h incubation. These data indicate that impairment of myelin structure and glutamatergic system and induction of oxidative stress are involved in the neuropathophysiology of NKH.


Subject(s)
Hyperglycinemia, Nonketotic , Humans , Animals , Rats , Hyperglycinemia, Nonketotic/genetics , Hyperglycinemia, Nonketotic/metabolism , Glycine , Myelin Sheath/metabolism , Oxidation-Reduction , Synaptic Transmission , Homeostasis
2.
Int J Mol Sci ; 22(19)2021 Oct 06.
Article in English | MEDLINE | ID: mdl-34639161

ABSTRACT

(1) Background: Calcium-binding protein S100B is involved in neuroregeneration but has also been associated with neurodegeneration. These contrasting effects may result from concentration or duration of exposure. We investigated the effect of long-term increased S100B levels on amyloid-ß processing in one-year-old transgenic (tg) mice with 12 copies of the murine S100B gene with specific consideration of sex and specific brain regions. (2) Methods: S100B and amyloid-ß 42 (Aß42) were quantified in serum, cerebrospinal fluid (CSF), adipose tissue, and different brain regions by ELISA in wild-type (wt) and S100Btg mice (each n = 7 per group). Thioflavin T (ThT) and Aß immunostaining were performed for visualization of Aß deposition. (3) Results: S100B in serum, CSF, and brain was significantly increased in S100Btg mice of both sexes. Aß42 was significantly increased in the hippocampus of male S100Btg mice (p = 0.0075), and the frontal cortex of female S100Btg mice (p = 0.0262). ThT and Aß immunostaining demonstrated Aß deposition in different brain regions in S100Btg mice of both sexes and female wt. (4) Conclusion: Our data validate this experimental model for studying the role of S100B in neurodegeneration and indicate that Aß processing is sex-dependent and brain region-specific, which deserves further investigation of signaling pathways and behavioral responses.


Subject(s)
Adipose Tissue/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/metabolism , Hippocampus/metabolism , Protein Processing, Post-Translational , S100 Calcium Binding Protein beta Subunit/metabolism , Alzheimer Disease/metabolism , Animals , Female , Male , Mice , Mice, Transgenic , S100 Calcium Binding Protein beta Subunit/genetics , Sex Factors
3.
Birth Defects Res ; 113(1): 22-31, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33009728

ABSTRACT

BACKGROUND: Zika virus (ZIKV) was confirmed to be related to microcephaly in 2016. However, there is still a need for understanding the embryonic morphological changes induced by ZIKV and when they occur. Here, chicken embryos were chosen as experimental model of ZIKV to evaluate virus-associated morphological alterations that might take place during embryonic development. METHODS: A screening with different viral doses was conducted in embryos at HH Stage 10-12 (E1.5) as well as a follow up of the first 5 days postinfection (dpi) was performed to observe the main morphologic changes post ZIKV infection. RESULTS: ZIKV exposed embryos presented a higher prevalence of mortality and defects such as brain malformation when compared to controls. Moreover, we observed that the phenotypes become more evident at 4dpi, when the viral load quantification reaches a peak. CONCLUSIONS: We found that ZIKV exposed embryos presented a high prevalence of mortality and central nervous system (CNS) abnormalities in a dose-dependent manner. The phenotype was more evident 4 days postinfection, when the viral load quantification reached a peak.


Subject(s)
Pregnancy Complications, Infectious , Zika Virus Infection , Zika Virus , Animals , Brain , Chick Embryo , Chickens , Female , Pregnancy
4.
Sleep Sci ; 13(1): 37-48, 2020.
Article in English | MEDLINE | ID: mdl-32670491

ABSTRACT

OBJECTIVE: To translate the Sleep Hygiene Index (SHI) to Brazilian Portuguese, to describe its psychometric properties and to show its association with sleep quality, daytime sleepiness, risk for sleep apnea and depressive symptoms. METHODS: Thirty subjects participated in the cultural adaptation and the item clarity evaluation. Twenty subjects answered the instrument in three different time-points for test-retest reliability. Eighty adult workers completed the SHI, the Pittsburgh Sleep Quality Index (PSQI), the Epworth Sleepiness Scale (ESS), the Beck Depression Inventory (BDI) and the STOP-BANG (S-B). RESULTS: SHI shows an acceptable internal consistency (Cronbach's α=0.75), as well as a high reproducibility (intraclass correlation=0.972, p<0.01). The three final factors of confirmatory factor analysis extract an average of 48.22% of the total sample variance. Worse sleep hygiene (higher SHI score) correlated with poor sleep quality (r=0.398, p<0.001), excessive daytime sleepiness (r=0.406, p<0.001) and depressive symptoms (r=0.324, p=0.003). No correlations with S-B were found. CONCLUSIONS: SHI presents satisfactory-to-optimal psychometric properties. This instrument is useful for treatment planning and management of sleep hygiene practices. Thus, it represents a reliable way of assessing sleep hygiene quantitatively in both research and clinical settings.

5.
Biomed Pharmacother ; 111: 1438-1446, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30841459

ABSTRACT

Mitochondria play an important role in cell life and in the regulation of cell death. In addition, mitochondrial dysfunction contributes to a wide range of neuropathologies. The nucleoside Guanosine (GUO) is an endogenous molecule, presenting antioxidant properties, possibly due to its direct scavenging ability and/or from its capacity to activate the antioxidant defense system. GUO demonstrate a neuroprotective effect due to the modulation of the glutamatergic system and maintenance of the redox system. Thus, considering the few studies focused on the direct effects of GUO on mitochondrial bioenergetics, we designed a study to evaluate the in vitro effects of GUO on rat mitochondrial function, as well as against Ca2+-induced impairment. Our results indicate that GUO prevented mitochondrial dysfunction induced by Ca2+ misbalance, once GUO was able to reduce mitochondrial swelling in the presence of Ca2+, as well as ROS production and hydrogen peroxide levels, and to increase manganese superoxide dismutase activity, oxidative phosphorylation and tricarboxylic acid cycle activities. Our study indicates for the first time that GUO could direct prevent the mitochondrial damage induced by Ca2+ and that these effects were not related to its scavenging properties. Our data indicates that GUO could be included as a new pharmacological strategy for diseases linked to mitochondrial dysfunction.


Subject(s)
Calcium/metabolism , Guanosine/pharmacology , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/metabolism , Neuroprotective Agents/pharmacology , Animals , Antioxidants/pharmacology , Citric Acid Cycle/drug effects , Hydrogen Peroxide/metabolism , Male , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Oxidative Phosphorylation/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
6.
Neurochem Res ; 43(7): 1476-1489, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29855847

ABSTRACT

Guanosine (GUO) has neuroprotective effects in experimental models of brain diseases involving glutamatergic excitotoxicity in male animals; however, its effects in female animals are poorly understood. Thus, we investigated the influence of gender and GUO treatment in adult male and female Wistar rats submitted to focal permanent cerebral ischemia in the motor cortex brain. Female rats were subdivided into non-estrogenic and estrogenic phase groups by estrous cycle verification. Immediately after surgeries, the ischemic animals were treated with GUO or a saline solution. Open field and elevated plus maze tasks were conducted with ischemic and naïve animals. Cylinder task, immunohistochemistry and infarct volume analyses were conducted only with ischemic animals. Female GUO groups achieved a full recovery of the forelimb symmetry at 28-35 days after the insult, while male GUO groups only partially recovered at 42 days, in the final evaluation. The ischemic insult affected long-term memory habituation to novelty only in female groups. Anxiety-like behavior, astrocyte morphology and infarct volume were not affected. Regardless the estrous cycle, the ischemic injury affected differently female and male animals. Thus, this study points that GUO is a potential neuroprotective compound in experimental stroke and that more studies, considering the estrous cycle, with both genders are recommended in future investigation concerning brain diseases.


Subject(s)
Brain Ischemia/prevention & control , Cerebral Cortex/drug effects , Guanosine/administration & dosage , Neuroprotective Agents/administration & dosage , Sex Characteristics , Animals , Brain Ischemia/pathology , Cerebral Cortex/pathology , Female , Male , Maze Learning/drug effects , Maze Learning/physiology , Rats , Rats, Wistar , Recovery of Function/drug effects , Recovery of Function/physiology
7.
Parasit Vectors ; 11(1): 191, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29554958

ABSTRACT

BACKGROUND: Cerebral malaria, the main complication of Plasmodium falciparum infection in humans, is associated with persistent neurocognitive sequels both in human disease and the murine experimental model. In recent years, cognitive deficits related to uncomplicated (non-cerebral) malaria have also been reported in chronically exposed residents of endemic areas, but not in some murine experimental models of non-cerebral malaria. This study aimed at evaluating the influence of uncomplicated malaria on different behavioural paradigms associated with memory and anxiety-like parameters in a murine model that has the ability to develop cerebral malaria. METHODS: Plasmodium berghei ANKA-infected and non-infected C57BL/6 mice were used. Development of cerebral malaria was prevented by chloroquine treatment starting on the fourth day of infection. The control group (non-infected mice) were treated with PBS. The effect of uncomplicated malaria infection on locomotor habituation, short and long-term memory and anxious-like behaviour was evaluated 64 days after parasite clearance in assays including open field, object recognition, Y-maze and light/dark tasks. RESULTS: Plasmodium berghei ANKA-infected mice showed significant long-lasting disturbances reflected by a long-term memory-related behaviour on open field and object recognition tasks, accompanied by an anxious-like phenotype availed on open field and light-dark tasks. CONCLUSIONS: Long-term neurocognitive sequels may follow an uncomplicated malaria episode in an experimental model prone to develop cerebral malaria, even if the infection is treated before the appearance of clinical signs of cerebral impairment.


Subject(s)
Anxiety , Malaria/complications , Memory , Time , Animals , Antimalarials/therapeutic use , Brain/parasitology , Cognition Disorders/etiology , Cognition Disorders/parasitology , Disease Models, Animal , Malaria/parasitology , Malaria, Cerebral , Mice , Mice, Inbred C57BL , Parasitemia/drug therapy , Plasmodium berghei/isolation & purification
8.
Epilepsia ; 58(10): 1771-1781, 2017 10.
Article in English | MEDLINE | ID: mdl-28762469

ABSTRACT

OBJECTIVES: Glutaric acidemia type I (GA-I) is an inherited neurometabolic disorder caused by deficiency of glutaryl-CoA dehydrogenase (GCDH) and characterized by increased levels of glutaric, 3-OH-glutaric, and glutaconic acids in the brain parenchyma. The increment of these organic acids inhibits glutamate decarboxylase (GAD) and consequently lowers the γ-aminobutyric acid (GABA) synthesis. Untreated patients exhibit severe neurologic deficits during development, including epilepsy, especially following an acute encephalopathy outbreak. In this work, we evaluated the role of the GABAergic system on epileptogenesis in GA-I using the Gcdh-/- mice exposed to a high lysine diet (Gcdh-/- -Lys). METHODS: Spontaneous recurrent seizures (SRS), seizure susceptibility, and changes in brain oscillations were evaluated by video-electroencephalography (EEG). Cortical GABAergic synaptic transmission was evaluated using electrophysiologic and neurochemical approaches. RESULTS: SRS were observed in 72% of Gcdh-/- -Lys mice, whereas no seizures were detected in age-matched controls (Gcdh+/+ or Gcdh-/- receiving normal diet). The severity and number of PTZ-induced seizures were higher in Gcdh-/- -Lys mice. EEG spectral analysis showed a significant decrease in theta and gamma oscillations and predominant delta waves in Gcdh-/- -Lys mice, associated with increased EEG left index. Analysis of cortical synaptosomes revealed a significantly increased percentage of glutamate release and decreased GABA release in Gcdh-/- -Lys mice that were associated with a decrease in cortical GAD immunocontent and activity and confirmed by reduced frequency of inhibitory events in cortical pyramidal cells. SIGNIFICANCE: Using an experimental model with a phenotype similar to that of GA-I in humans-the Gcdh-/- mice under high lysine diet (Gcdh-/- -Lys)-we provide evidence that a reduction in cortical inhibition of Gcdh-/- -Lys mice, probably induced by GAD dysfunction, leads to hyperexcitability and increased slow oscillations associated with neurologic abnormalities in GA-I. Our findings offer a new perspective on the pathophysiology of brain damage in GA-I.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/genetics , Brain/drug effects , Epilepsy/genetics , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , gamma-Aminobutyric Acid/drug effects , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Blotting, Western , Brain Diseases, Metabolic/metabolism , Chromatography, High Pressure Liquid , Epilepsy/metabolism , GABA Antagonists/pharmacology , Glutamate Decarboxylase , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Mice , Mice, Knockout , Pentylenetetrazole/pharmacology , Synaptosomes/drug effects , Synaptosomes/metabolism , gamma-Aminobutyric Acid/metabolism
9.
Neurotox Res ; 32(2): 276-290, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28429309

ABSTRACT

Tissue accumulation of α-ketoadipic (KAA) and α-aminoadipic (AAA) acids is the biochemical hallmark of α-ketoadipic aciduria. This inborn error of metabolism is currently considered a biochemical phenotype with uncertain clinical significance. Considering that KAA and AAA are structurally similar to α-ketoglutarate and glutamate, respectively, we investigated the in vitro effects of these compounds on glutamatergic neurotransmission in the brain of adolescent rats. Bioenergetics and redox homeostasis were also investigated because they represent fundamental systems for brain development and functioning. We first observed that AAA significantly decreased glutamate uptake, whereas glutamate dehydrogenase activity was markedly inhibited by KAA in a competitive fashion. In addition, AAA and more markedly KAA induced generation of reactive oxygen and nitrogen species (increase of 2',7'-dichloroflurescein (DCFH) oxidation and nitrite/nitrate levels), lipid peroxidation (increase of malondialdehyde concentrations), and protein oxidation (increase of carbonyl formation and decrease of sulfhydryl content), besides decreasing the antioxidant defenses (reduced glutathione (GSH)) and aconitase activity. Furthermore, KAA-induced lipid peroxidation and GSH decrease were prevented by the antioxidants α-tocopherol, melatonin, and resveratrol, suggesting the involvement of reactive species in these effects. Noteworthy, the classical inhibitor of NMDA glutamate receptors MK-801 was not able to prevent KAA-induced and AAA-induced oxidative stress, determined by DCFH oxidation and GSH levels, making unlikely a secondary induction of oxidative stress through overstimulation of glutamate receptors. In contrast, KAA and AAA did not significantly change brain bioenergetic parameters. We speculate that disturbance of glutamatergic neurotransmission and redox homeostasis by KAA and AAA may play a role in those cases of α-ketoadipic aciduria that display neurological symptoms.


Subject(s)
2-Aminoadipic Acid/pharmacology , Adipates/pharmacology , Cerebral Cortex/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Oxidative Stress/drug effects , Adenosine Triphosphatases/metabolism , Animals , Cell Membrane/drug effects , Glutamate Dehydrogenase/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamic Acid/metabolism , Homeostasis/drug effects , Liver/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Multienzyme Complexes/metabolism , Protein Carbonylation/drug effects , Rats , Synapses/drug effects , Tritium/metabolism
10.
Eur Neuropsychopharmacol ; 26(2): 368-377, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26792443

ABSTRACT

Attention deficit hyperactivity disorder (ADHD) is characterized by impairing levels of hyperactivity, impulsivity and inattention. However, different meta-analyses have reported disruptions in short and long-term memory in ADHD patients. Previous studies indicate that mnemonic dysfunctions might be the result of deficits in attentional circuits, probably due to ineffective dopaminergic modulation of hippocampal synaptic plasticity. In this study we aimed to evaluate the potential therapeutic effects of a neuromodulatory technique, transcranial direct current stimulation (tDCS), in short-term memory (STM) deficits presented by the spontaneous hypertensive rats (SHR), the most widely used animal model of ADHD. Adult male SHR and Wistar Kyoto rats (WKY) were subjected to a constant electrical current of 0.5 mA intensity applied on the frontal cortex for 20 min/day during 8 days. STM was evaluated with an object recognition test conducted in an open field. Exploration time and locomotion were recorded, and brain regions were dissected to determine dopamine and BDNF levels. SHR spent less time exploring the new object when compared to WKY, and tDCS improved object recognition deficits in SHR without affecting WKY performance. Locomotor activity was higher in SHR and it was not affected by tDCS. After stimulation, dopamine levels were increased in the hippocampus and striatum of both strains, while BDNF levels were increased only in the striatum of WKY. These findings suggest that tDCS on the frontal cortex might be able to improve STM deficits present in SHR, which is potentially related to dopaminergic neurotransmission in the hippocampus and striatum of those animals.


Subject(s)
Attention Deficit Disorder with Hyperactivity/complications , Memory Disorders/etiology , Memory Disorders/therapy , Memory, Short-Term/physiology , Transcranial Direct Current Stimulation , Analysis of Variance , Animals , Attention Deficit Disorder with Hyperactivity/pathology , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Dopamine/metabolism , Locomotion/physiology , Male , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Recognition, Psychology/physiology , Statistics as Topic
11.
J Neurol Sci ; 359(1-2): 133-40, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26671102

ABSTRACT

Glutaric aciduria type I (GA I) is biochemically characterized by accumulation of glutaric and 3-hydroxyglutaric acids in body fluids and tissues, particularly in the brain. Affected patients show progressive cortical leukoencephalopathy and chronic degeneration of the basal ganglia whose pathogenesis is still unclear. In the present work we investigated parameters of bioenergetics and redox homeostasis in various cerebral structures (cerebral cortex, striatum and hippocampus) and heart of adult wild type (Gcdh(+/+)) and glutaryl-CoA dehydrogenase deficient knockout (Gcdh(-/-)) mice fed a baseline chow. Oxidative stress parameters were also measured after acute lysine overload. Finally, mRNA expression of NMDA subunits and GLT1 transporter was determined in cerebral cortex and striatum of these animals fed a baseline or high lysine (4.7%) chow. No significant alterations of bioenergetics or redox status were observed in these mice. In contrast, mRNA expression of the NR2B glutamate receptor subunit and of the GLT1 glutamate transporter was higher in cerebral cortex of Gcdh(-/-) mice. Furthermore, NR2B expression was markedly elevated in striatum of Gcdh(-/-) animals receiving chronic Lys overload. These data indicate higher susceptibility of Gcdh(-/-) mice to excitotoxic damage, implying that this pathomechanism may contribute to the cortical and striatum alterations observed in GA I patients.


Subject(s)
Amino Acid Metabolism, Inborn Errors/complications , Brain Diseases, Metabolic/complications , Brain Injuries/etiology , Gene Expression Regulation/genetics , Glutaryl-CoA Dehydrogenase/deficiency , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Catalase/metabolism , Disease Models, Animal , Excitatory Amino Acid Transporter 2/metabolism , Fluoresceins/metabolism , Glucosephosphate Dehydrogenase/metabolism , Glutaryl-CoA Dehydrogenase/genetics , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Malondialdehyde/metabolism , Mice , Mice, Transgenic , NAD/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Sulfhydryl Compounds/metabolism , Superoxide Dismutase/metabolism
12.
Mol Neurobiol ; 52(1): 710-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25280668

ABSTRACT

The 14-3-3 protein family takes part in a wide range of cellular processes and is expressed in all eukaryotic organisms. In mammals, seven isoforms (ß, ε, η, γ, τ, ζ, and σ) have been identified. 14-3-3 proteins are suggested to modulate the insulin-signaling cascade in the brain. The aim of this study was to investigate whether insulin resistance state induced by high palatable diet modulates expression of the 14-3-3 proteins in brain. Wistar male rats (n = 8) were divided into two experimental groups: insulin resistant (IR), induced by high palatable diet, and control (CO) group. Biochemical parameters (glucose tolerance test and plasma lipid profile) were evaluated after 130 days. Brain structures (cortex and hippocampus) were dissected for evaluation of messenger RNA (mRNA) and protein levels of different 14-3-3 proteins. Statistical analyses included Student t test and Pearson correlation. Significant decrease was observed in Ywhah and in Ywahq mRNA levels in the cortex of IR group, while no changes were observed in the hippocampus. Significant increase of θ isoform was observed in hippocampus IR group by immunodetection, while no differences were detected in the remaining isoforms. Inverse correlation was observed between blood glucose levels in cortex IR group and both Ywhah and Ywhaq mRNA levels. Protein levels of Creb and phosphatidylinositide 3-kinases (PI3K) showed to be increased in the hippocampus. These alterations may be due to a compensatory effect of impaired insulin signaling. We demonstrated differential expression of 14-3-3 isoforms throughout brain regions of rats with IR. As a whole, our results indicate that brain 14-3-3 levels are influenced by different diets.


Subject(s)
14-3-3 Proteins/metabolism , Brain/metabolism , Diet , Insulin Resistance , 14-3-3 Proteins/genetics , Animals , Blood Glucose/metabolism , Blotting, Western , Cerebral Cortex/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Disease Models, Animal , Glucose Tolerance Test , Hippocampus/metabolism , Lipids/blood , Male , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar
13.
J Neurol Sci ; 346(1-2): 260-7, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25241940

ABSTRACT

The role of excitotoxicity on the neuropathology of glutaric acidemia type I (GA I) is still under debate. Therefore, in the present work, we evaluated glutamate uptake by brain slices and glutamate binding to synaptic membranes, as well as glutamine synthetase activity in cerebral cortex and striatum from glutaryl-CoA dehydrogenase deficient (Gcdh(-/-)) mice along development (7, 15, 30 and 60 days of life) in the hopes of clarifying this matter. We also tested the influence of glutaric acid (GA) added exogenously on these parameters. [(3)H]Glutamate uptake was not significantly altered in cerebral cortex and striatum from Gcdh(-/-) mice, as compared to WT mice. However, GA provoked a significant decrease of [(3)H]glutamate uptake in striatum from both WT and Gcdh(-/-) mice older than 7 days. This inhibitory effect was more pronounced in Gcdh(-/-), as compared to WT mice. The use of a competitive inhibitor of glutamate astrocytic transporters indicated that the decrease of [(3)H]glutamate uptake caused by GA was due to the competition between this organic acid and glutamate for the same astrocytic transporter site. We also found that Na(+)-dependent [(3)H]glutamate binding (binding to transporters) was increased in the striatum from Gcdh(-/-) mice and that GA significantly diminished this binding both in striatum and cerebral cortex from Gcdh(-/-), but not from WT mice. Finally, we observed that glutamine synthetase activity was not changed in brain cortex and striatum from Gcdh(-/-) and WT mice and that GA was not able to alter this activity. It is therefore presumed that a disturbance of the glutamatergic neurotransmission system caused by GA may potentially be involved in the neuropathology of GA I, particularly in the striatum.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic/metabolism , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Glutarates/pharmacology , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Brain Diseases, Metabolic/pathology , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Corpus Striatum/drug effects , Corpus Striatum/pathology , Disease Models, Animal , Glutamate-Ammonia Ligase/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Mice , Mice, Knockout
14.
Cerebellum ; 13(6): 751-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25172216

ABSTRACT

Zellweger syndrome (ZS) and some peroxisomal diseases are severe inherited disorders mainly characterized by neurological symptoms and cerebellum abnormalities, whose pathogenesis is poorly understood. Biochemically, these diseases are mainly characterized by accumulation of pristanic acid (Prist) and other fatty acids in the brain and other tissues. In this work, we evaluated the in vitro influence of Prist on redox homeostasis by measuring lipid, protein, and DNA damage, as well as the antioxidant defenses and the activities of aconitase and α-ketoglutarate dehydrogenase in cerebellum of 30-day-old rats. The effect of Prist on DNA damage was also evaluated in blood of these animals. Some parameters were also evaluated in cerebellum from neonatal rats and in cerebellum neuronal cultures. Prist significantly increased malondialdehyde (MDA) levels and carbonyl formation and reduced sulfhydryl content and glutathione (GSH) concentrations in cerebellum of young rats. It also caused DNA strand damage in cerebellum and induced a high micronuclei frequency in blood. On the other hand, this fatty acid significantly reduced α-ketoglutarate dehydrogenase and aconitase activities in rat cerebellum. We also verified that Prist-induced increase of MDA levels was totally prevented by melatonin and attenuated by α-tocopherol but not by the nitric oxide synthase inhibitor N(ω)-nitro-L-arginine methyl ester, indicating the involvement of reactive oxygen species in this effect. Cerebellum from neonate rats also showed marked alterations of redox homeostasis, including an increase of MDA levels and a decrease of sulfhydryl content and GSH concentrations elicited by Prist. Finally, Prist provoked an increase of dichlorofluorescein (DCFH) oxidation in cerebellum-cultivated neurons. Our present data indicate that Prist compromises redox homeostasis in rat cerebellum and blood and inhibits critical enzymes of the citric acid cycle that are susceptible to free radical attack. The present findings may contribute to clarify the pathogenesis of the cerebellar alterations observed in patients affected by ZS and some peroxisomal disorders in which Prist is accumulated.


Subject(s)
Antioxidants/metabolism , Cerebellum/drug effects , Cerebellum/metabolism , Fatty Acids/toxicity , Oxidation-Reduction/drug effects , Aconitate Hydratase/metabolism , Animals , Animals, Newborn , Cells, Cultured , DNA Damage/drug effects , Fluoresceins/metabolism , Glutathione/metabolism , Homeostasis/drug effects , Ketoglutarate Dehydrogenase Complex/metabolism , Malondialdehyde/metabolism , Melatonin/administration & dosage , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/administration & dosage , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Rats, Wistar , Sulfhydryl Compounds/metabolism , alpha-Tocopherol/pharmacology
15.
Neurochem Res ; 39(5): 973-84, 2014 May.
Article in English | MEDLINE | ID: mdl-24706093

ABSTRACT

Several researchers have recently used C6 cells to evaluate functional properties of high-affinity glutamate transporters. However, it has been demonstrated that this lineage suffers several morphological and biochemical alterations according to the number of passages in culture. Currently, there are no reports showing whether functional properties of high-affinity glutamate transporters comply with these sub culturing-dependent modifications. The present study aimed to compare the functional properties of high-affinity glutamate transporters expressed in early (EPC6) and late (LPC6) passage C6 cells through a detailed pharmacological and biochemical characterization. Between 60-180 min of L-[(3)H]glu incubation, LPC6 presented an intracellular [(3)H] 55% lower than EPC6. Both cultures showed a time-dependent increase of intracellular [(3)H] reaching maximal levels at 120 min. Cultures incubated with D-[(3)H]asp showed a time-dependent increase of [(3)H] until 180 min. Moreover, LPC6 have a D-[(3)H]asp-derived intracellular [(3)H] 30-45% lower than EPC6 until 120 min. Only EAAT3 was immunodetected in cultures and its total content was equal between them. PMA-stimulated EAAT3 trafficking to membrane increased 50% of L-[(3)H]glu-derived intracellular [(3)H] in EPC6 and had no effect in LPC6. LPC6 displayed characteristics that resemble senescence, such as high ß-Gal staining, cell enlargement and increase of large and regular nuclei. Our results demonstrated that LPC6 exhibited glutamate uptake impairment, which may have occurred due to its inability to mobilize EAAT3 to cell membrane. This profile might be related to senescent process observed in this culture. Our results suggest that LPC6 cells are an inappropriate glial cellular model to investigate the functional properties of high-affinity glutamate transporters.


Subject(s)
Aspartic Acid/metabolism , Cellular Senescence/physiology , Glutamic Acid/metabolism , Animals , Glioma/metabolism , Rats, Wistar , Tritium , Tumor Cells, Cultured
16.
PLoS One ; 9(4): e94581, 2014.
Article in English | MEDLINE | ID: mdl-24733087

ABSTRACT

BACKGROUND: Early adverse experiences are associated with increased risk of developing psychiatric disorders, although little is known about the neurobiological mediators involved. The mechanisms by which early environmental influences may mediate vulnerability in the development of offspring await further investigation. The present study correlated the NGF, BDNF, IL-6 and cortisol levels of mothers with postpartum affective disorders (PPAD) with infant development. METHODS: A longitudinal study was performed with 152 pregnant women and their infants. Between 60 and 120 days after delivery, women were interviewed and provided biological samples for biochemical analysis, and the infants were examined for neurobiological-motor development. RESULTS: Overall, the mothers' history of affective disorders, PPAD and anxiety disorder were associated with infant motor development. Using an adjusted linear regression analysis, PPAD (p = 0.049), maternal anxiety disorder (p = 0.043), NGF level (p = 0.034) and infant cortisol level (p = 0.013) were associated with infant motor development. Using a factorial analysis of primary components, two components were retained. The psychological factor was characterized by a positive loading of a history of affective disorder, PPAD and anxiety disorder. For the biological factor, infant cortisol adhered negatively with infant motor development, but NGF was positively associated. The psychological factor had a negative association, but the biological factor had a positive association with infant motor development. CONCLUSIONS: There are few studies that have focused on the relationship of biomarkers and infant neurodevelopment. Our study points that psychological and biological factors are associated with infant motor development, however the causal relationship between these factors is still to be defined.


Subject(s)
Brain-Derived Neurotrophic Factor/blood , Interleukin-6/blood , Mood Disorders/blood , Nerve Growth Factor/blood , Adult , Anxiety Disorders/blood , Anxiety Disorders/physiopathology , Female , Humans , Infant , Infant, Newborn , Longitudinal Studies , Male , Mental Disorders/blood , Mental Disorders/physiopathology , Mood Disorders/physiopathology , Mothers/psychology , Motor Skills , Postpartum Period , Pregnancy , Pregnancy Complications , Principal Component Analysis , Puerperal Disorders/blood , Puerperal Disorders/physiopathology
17.
PLoS One ; 9(3): e90477, 2014.
Article in English | MEDLINE | ID: mdl-24594605

ABSTRACT

We determined mRNA expression of the ionotropic glutamate receptors NMDA (NR1, NR2A and NR2B subunits), AMPA (GluR2 subunit) and kainate (GluR6 subunit), as well as of the glutamate transporters GLAST and GLT1 in cerebral cortex and striatum of wild type (WT) and glutaryl-CoA dehydrogenase deficient (Gchh-/-) mice aged 7, 30 and 60 days. The protein expression levels of some of these membrane proteins were also measured. Overexpression of NR2A and NR2B in striatum and of GluR2 and GluR6 in cerebral cortex was observed in 7-day-old Gcdh-/-. There was also an increase of mRNA expression of all NMDA subunits in cerebral cortex and of NR2A and NR2B in striatum of 30-day-old Gcdh-/- mice. At 60 days of life, all ionotropic receptors were overexpressed in cerebral cortex and striatum of Gcdh-/- mice. Higher expression of GLAST and GLT1 transporters was also verified in cerebral cortex and striatum of Gcdh-/- mice aged 30 and 60 days, whereas at 7 days of life GLAST was overexpressed only in striatum from this mutant mice. Furthermore, high lysine intake induced mRNA overexpression of NR2A, NR2B and GLAST transcripts in striatum, as well as of GluR2 and GluR6 in both striatum and cerebral cortex of Gcdh-/- mice. Finally, we found that the protein expression of NR2A, NR2B, GLT1 and GLAST were significantly greater in cerebral cortex of Gcdh-/- mice, whereas NR2B and GLT1 was similarly enhanced in striatum, implying that these transcripts were translated into their products. These results provide evidence that glutamate receptor and transporter expression is higher in Gcdh-/- mice and that these alterations may be involved in the pathophysiology of GA I and possibly explain, at least in part, the vulnerability of striatum and cerebral cortex to injury in patients affected by GA I.


Subject(s)
Amino Acid Metabolism, Inborn Errors/pathology , Amino Acid Transport System X-AG/metabolism , Brain Diseases, Metabolic/pathology , Cerebral Cortex/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Neostriatum/metabolism , Receptors, Glutamate/metabolism , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Transport System X-AG/genetics , Animals , Brain Diseases, Metabolic/enzymology , Cerebral Cortex/pathology , Diet , Female , Gene Expression Regulation , Glutaryl-CoA Dehydrogenase/metabolism , Lysine/metabolism , Male , Mice , Neostriatum/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Glutamate/genetics
18.
Mov Disord ; 29(4): 568-73, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24399647

ABSTRACT

BACKGROUND: Because lithium exerts neuroprotective effects in preclinical models of polyglutamine disorders, our objective was to assess the safety and efficacy of lithium carbonate (0.5-0.8 milliequivalents per liter) in patients with Machado-Joseph disease (spinocerebellar ataxia type 3 [MJD/SCA3]). METHODS: For this phase 2, single-center, double-blind, parallel, placebo-controlled trial (ClinicalTrials.gov identifier NCT01096082), 62 patients who had MJD/SCA3 with a disease duration ≤10 years and an independent gait were randomly assigned (1:1) to receive either lithium or placebo. RESULTS: After 24 weeks, 169 adverse events were reported, including 50.3% in the lithium group (P = 1.00; primary safety outcome). Sixty patients (31 in the placebo group and 29 in the lithium group) were analyzed for efficacy (intention-to-treat analysis). Mean progression between groups did not differ according to scores on the Neurological Examination Score for the Assessment of Spinocerebellar Ataxia (NESSCA) after 48 weeks (-0.35; 95% confidence interval, -1.7 to 1.0; primary efficacy outcome). The lithium group exhibited minor progression on the PATA speech-rate (P = 0.002), the nondominant Click Test (P = 0.023), the Spinocerebellar Ataxia Functional Index (P = 0.003), and the Composite Cerebellar Functional Score (P = 0.029). CONCLUSIONS: Lithium was safe and well tolerated, but it had no effect on progression when measured using the NESSCA in patients with MJD/SCA3. This slowdown in secondary outcomes deserves further clarification.


Subject(s)
Enzyme Inhibitors/therapeutic use , Lithium Carbonate/therapeutic use , Machado-Joseph Disease/drug therapy , Adult , Double-Blind Method , Enzyme Inhibitors/adverse effects , Female , Humans , Lithium Carbonate/adverse effects , Male , Middle Aged , Treatment Outcome
19.
Neurochem Res ; 38(11): 2359-63, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24013888

ABSTRACT

Neuropathological hallmarks of Alzheimer's disease (AD) include amyloid plaque formation, neurofibrillary tangles, neuronal and synaptic loss. This study aims to identify the neuroprotective effects of the selenium compounds on the neurotoxicity of amyloid ß(1-42) in primary cultures of murine hippocampal neurons. Samples were subjected to immunocytochemistry and western blotting techniques to determine the role of treatments on neuronal viability and synaptic protein SNAP-25. We observed a reduced cell viability amyloid ß-peptide (1-42)-induced. When cells were co-treated with amyloid ß-peptide (1-42) and selenium compounds, we verified a strong increase in relative cell viability and in the level of synaptic marker synaptosomal-associated protein SNAP-25 induced by selenium compounds.


Subject(s)
Amyloid beta-Peptides/toxicity , Azoles/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Organoselenium Compounds/pharmacology , Peptide Fragments/toxicity , Animals , Cell Survival/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Isoindoles , Rats , Synaptosomal-Associated Protein 25/metabolism
20.
Exp Neurol ; 247: 66-72, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23499835

ABSTRACT

The mitochondrial electron transport system (ETS) is a main source of cellular ROS, including hydrogen peroxide (H2O2). The production of H2O2 also involves the mitochondrial membrane potential (ΔΨm) and oxygen consumption. Impaired insulin signaling causes oxidative neuronal damage and places the brain at risk of neurodegeneration. We evaluated whether insulin signaling cross-talks with ETS components (complexes I and F0F1ATP synthase) and ΔΨm to regulate mitochondrial H2O2 production, in tissue preparations from rat brain. Insulin (50 to 100 ng/mL) decreased H2O2 production in synaptosomal preparations in high Na(+) buffer (polarized state), stimulated by glucose and pyruvate, without affecting the oxygen consumption. In addition, insulin (10 to 100 ng/mL) decreased H2O2 production induced by succinate in synaptosomes in high K(+) (depolarized state), whereas wortmannin and LY290042, inhibitors of the PI3K pathway, reversed this effect; heated insulin had no effect. Insulin decreased H2O2 production when complexes I and F0F1ATP synthase were inhibited by rotenone and oligomycin respectively suggesting a target effect on complex III. Also, insulin prevented the generation of maximum level of ∆Ψm induced by succinate. The PI3K inhibitors and heated insulin maintained the maximum level of ∆Ψm induced by succinate in synaptosomes in a depolarized state. Similarly, insulin decreased ROS production in neuronal cultures. In mitochondrial preparations, insulin neither modulated H2O2 production or oxygen consumption. In conclusion, the normal downstream insulin receptor signaling is necessary to regulate complex III of ETS avoiding the generation of maximal ∆Ψm and increased mitochondrial H2O2 production.


Subject(s)
Brain/ultrastructure , Hydrogen Peroxide/pharmacology , Insulin/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Oxidants/pharmacology , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/pharmacology , Animals , Cerebral Cortex/cytology , Dose-Response Relationship, Drug , Electron Transport , Embryo, Mammalian , Gene Expression Regulation/drug effects , Glutamic Acid/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Neurons/drug effects , Neurons/metabolism , Oxygen Consumption , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Synaptosomes/drug effects , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...