Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Neurotox Res ; 29(3): 408-18, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26694914

ABSTRACT

Severe hyperhomocysteinemia is caused by increased plasma levels of homocysteine (Hcy), a methionine derivative, and is associated with cerebral disorders. Creatine supplementation has emerged as an adjuvant to protect against neurodegenerative diseases, due to its potential antioxidant role. Here, we examined the effects of severe hyperhomocysteinemia on brain metabolism, and evaluated a possible neuroprotective role of creatine in hyperhomocysteinemia, by concomitant treatment with Hcy and creatine (50 mg/Kg body weight). Hyperhomocysteinemia was induced in young rats (6-day-old) by treatment with homocysteine (0.3-0.6 µmol/g body weight) for 23 days, and then the following parameters of rat amygdala were evaluated: (1) the activity of the respiratory chain complexes succinate dehydrogenase, complex II and cytochrome c oxidase; (2) mitochondrial mass and membrane potential; (3) the levels of necrosis and apoptosis; and (4) the activity and immunocontent of Na(+),K(+)-ATPase. Hcy treatment decreased the activities of succinate dehydrogenase and cytochrome c oxidase, but did not alter complex II activity. Hcy treatment also increased the number of cells with high mitochondrial mass, high mitochondrial membrane potential, and in late apoptosis. Importantly, creatine administration prevented some of the key effects of Hcy administration on the amygdala. We also observed a decrease in the activity and immunocontent of the α1 subunit of the Na(+),K(+)-ATPase in amygdala after Hcy- treatment. Our findings support the notion that Hcy modulates mitochondrial function and bioenergetics in the brain, as well as Na(+),K(+)-ATPase activity, and suggest that creatine might represent an effective adjuvant to protect against the effects of high Hcy plasma levels.


Subject(s)
Amygdala/metabolism , Creatine/administration & dosage , Electron Transport Chain Complex Proteins/metabolism , Hyperhomocysteinemia/metabolism , Mitochondria/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Amygdala/drug effects , Animals , Apoptosis/drug effects , Electron Transport Chain Complex Proteins/drug effects , Electron Transport Complex IV/metabolism , Female , Homocysteine/blood , Homocysteine/toxicity , Hyperhomocysteinemia/chemically induced , Male , Mitochondria/drug effects , Necrosis/chemically induced , Rats , Rats, Wistar , Succinate Dehydrogenase/metabolism
2.
Toxicol In Vitro ; 29(8): 2022-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26318273

ABSTRACT

Ammonia is a key toxin in the precipitation of hepatic encephalopathy (HE), a neuropsychiatric disorder associated with liver failure. In response to ammonia, various toxic events are triggered in astroglial cells, and alterations in brain glutamate communication are common. Resveratrol is a polyphenolic compound that has been extensively studied in pathological events because it presents several beneficial effects, including some in the central nervous system (CNS). We previously described that resveratrol is able to significantly modulate glial functioning and has a protective effect during ammonia challenge in vitro. In this study, we addressed the mechanisms by which resveratrol can protect C6 astroglial cells from glutamatergic alterations induced by ammonia. Resveratrol was able to prevent all the effects triggered by ammonia: (i) decrease in glutamate uptake activity and expression of the EAAC1 glutamate transporter, the main glutamate transporter present in C6 cells; (ii) increase of glutamate release, which was also dependent on the activation of the Na(+)-K(+)-Cl(-) co-transporter NKCC1; (iii) reduction in GS activity and intracellular GSH content; and (iv) impairment of Na(+)K(+)-ATPase activity. Interestingly, resveratrol, per se, also positively modulated the astroglial functions evaluated. Moreover, we demonstrated that heme oxygenase 1 (HO1), an enzyme that is part of the cellular defense system, mediated some of the effects of resveratrol. In conclusion, the mechanisms of the putative protective role of resveratrol against ammonia toxicity involve the modulation of pathways and molecules related to glutamate communication in astroglial cells.


Subject(s)
Ammonia/toxicity , Astrocytes/drug effects , Glutamic Acid/metabolism , Stilbenes/pharmacology , Animals , Astrocytes/metabolism , Cell Line, Tumor , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Gene Expression Regulation/drug effects , Glutamate-Ammonia Ligase/metabolism , Glutathione , Rats , Resveratrol , Signal Transduction , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism
3.
Metab Brain Dis ; 29(3): 825-35, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24810635

ABSTRACT

The reduction in the secretion of ovarian hormones, principally estrogen, is a consequence of menopause. Estrogens act primarily as female sex hormones, but also exert effects on different physiological systems including the central nervous system. The treatment normally used to reduce the symptoms of menopause is the hormone therapy, which seems to be effective in treating symptoms, but it may be responsible for adverse effects. Based on this, there is an increasing demand for alternative therapies that minimize signs and symptoms of menopause. In the present study we investigated the effect of ovariectomy and/or physical exercise on the activities of energy metabolism enzymes, such as creatine kinase (cytosolic and mitochondrial fractions), pyruvate kinase, succinate dehydrogenase, complex II, cytochrome c oxidase, as well as on ATP levels in the hippocampus of adult rats. Adult female Wistar rats with 90 days of age were subjected to ovariectomy (an animal model widely used to mimic the postmenopausal changes). Thirty days after the procedure, the rats were submitted to the exercise protocol, which was performed three times a week for 30 days. Twelve hours after the last training session, the rats were decapitated for subsequent biochemical analyzes. Results showed that ovariectomy did not affect the activities of pyruvate kinase, succinate dehydrogenase and complex II, but decreased the activities of creatine kinase (cytosolic and mitochondrial fractions) and cytochrome c oxidase. ATP levels were also reduced. Exercise did not produce the expected results since it was only able to partially reverse the activity of creatine kinase cytosolic fraction. The results of this study suggest that estrogen deficiency, which occurs as a result of ovariectomy, affects generation systems and energy homeostasis, reducing ATP levels in hippocampus of adult female rats.


Subject(s)
Adenosine Triphosphate/metabolism , Creatine Kinase/metabolism , Electron Transport Complex IV/metabolism , Hippocampus/metabolism , Ovariectomy , Physical Conditioning, Animal/physiology , Animals , Female , Pyruvate Kinase/metabolism , Rats , Rats, Wistar , Succinate Dehydrogenase/metabolism
4.
Gene ; 545(1): 72-9, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24797612

ABSTRACT

Homocystinuria is a neurometabolic disease caused by severe deficiency of cystathionine beta-synthase activity, resulting in severe hyperhomocysteinemia. Affected patients present several symptoms including a variable degree of motor dysfunction, being that the pathomechanism is not fully understood. In the present study we investigated the effect of chronic hyperhomocysteinemia on some parameters of oxidative stress, namely 2'7'dichlorofluorescein (DCFH) oxidation, levels of thiobarbituric acid-reactive substances (TBARS), antioxidant enzyme activities (SOD, CAT and GPx), reduced glutathione (GSH), total sulfhydryl and carbonyl content, as well as nitrite levels in soleus skeletal muscle of young rats subjected to model of severe hyperhomocysteinemia. We also evaluated the effect of creatine on biochemical alterations elicited by hyperhomocysteinemia. Wistar rats received daily subcutaneous injection of homocysteine (0.3-0.6 µmol/g body weight), and/or creatine (50mg/kg body weight) from their 6th to the 28th days age. Controls and treated rats were decapitated at 12h after the last injection. Chronic homocysteine administration increased 2'7'dichlorofluorescein (DCFH) oxidation, an index of production of reactive species and TBARS levels, an index of lipoperoxidation. Antioxidant enzyme activities, such as SOD and CAT were also increased, but GPx activity was not altered. The content of GSH, sulfhydril and carbonyl were decreased, as well as levels of nitrite. Creatine concurrent administration prevented some homocysteine effects probably by its antioxidant properties. Our data suggest that the oxidative insult elicited by chronic hyperhomocystenemia may provide insights into the mechanisms by which homocysteine exerts its effects on skeletal muscle function. Creatine prevents some alterations caused by homocysteine.


Subject(s)
Creatine/pharmacology , Homocysteine/pharmacology , Hyperhomocysteinemia/metabolism , Muscle, Skeletal/metabolism , Animals , Catalase/metabolism , Creatine/metabolism , Female , Fluoresceins/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Homeostasis/drug effects , Hyperhomocysteinemia/drug therapy , Male , Muscle, Skeletal/drug effects , Nitrites/metabolism , Oxidation-Reduction , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
5.
Nutr Neurosci ; 17(3): 127-37, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24621057

ABSTRACT

Previous studies have demonstrated that early environmental interventions influence the consumption of palatable food and the abdominal fat deposition in female rats chronically exposed to a highly caloric diet in adulthood. In this study, we verified the metabolic effects of chronic exposure to a highly palatable diet, and determine the response to its withdrawal in adult neonatally handled and non-handled rats. Consumption of foods (standard lab chow and chocolate), body weight gain, abdominal fat deposition, plasma triglycerides, and leptin, as well as serum butyrylcholinesterase (BuChE), and cerebral acetylcholinesterase (AChE) activities were measured during chronic chocolate exposure and after deprivation of this palatable food in female rats exposed or not to neonatal handling (10 minutes/day, 10 first days of life). Handled rats increased rebound chocolate consumption in comparison to non-handled animals after 1 week of chocolate withdrawal; these animals also decreased body weight in the first 24 hours but this effect disappeared after 7 days of withdrawal. Chocolate increased abdominal fat in non-handled females, and this effect remained after 30 days of withdrawal; no differences in plasma leptin were seen after 7 days of withdrawal. Chocolate also increased serum BuChE activity in non-handled females, this effect was still evident after 7 days of withdrawal, but it disappeared after 30 days of withdrawal. Chocolate deprivation decreased cerebral AChE activity in both handled and non-handled animals. These findings suggest that neonatal handling modulates the preference for palatable food and induces a specific metabolic response that may be more adaptive in comparison to non-handled rats.


Subject(s)
Animals, Newborn/physiology , Behavior, Animal , Diet , Environment , Handling, Psychological , Abdominal Fat , Acetylcholinesterase/metabolism , Adaptation, Psychological , Animals , Brain/enzymology , Butyrylcholinesterase/blood , Cacao , Energy Intake , Feeding Behavior/psychology , Female , Food Preferences/psychology , Leptin/blood , Obesity, Abdominal/etiology , Obesity, Abdominal/psychology , Pregnancy , Rats , Rats, Wistar , Stress, Psychological , Substance Withdrawal Syndrome , Triglycerides/blood , Weight Gain
6.
Mol Neurobiol ; 50(2): 589-96, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24590316

ABSTRACT

Mild hyperhomocysteinemia is considered to be a risk factor for cerebral and cardiovascular disorders and can be modeled in experimental rats. Inflammation has been implicated in the toxic effects of homocysteine. Cholinergic signaling controls cytokine production and inflammation through the "cholinergic anti-inflammatory pathway," and brain acetylcholinesterase activity plays a role in this regulation. The aim of this present study is to investigate the effect of mild chronic hyperhomocysteinemia on proinflammatory cytokine levels in the brain, heart, and serum of rats. Activity, immunocontent, and gene expression of acetylcholinesterase in the brain and butyrylcholinesterase activity in serum were also evaluated. Mild hyperhomocysteinemia was induced in Wistar rats by homocysteine administration (0.03 µmol/g of body weight) twice a day, from the 30th to the 60th days of life. Controls received saline in the same volumes. Results demonstrated an increase in tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and the chemokine monocyte chemotactic protein-1 (MCP-1) in the hippocampus, as well as an increase in IL-1ß and IL-6 levels in cerebral cortex. Acetylcholinesterase activity was increased in rats subjected to mild hyperhomocysteinemia in both cerebral structures tested; the immunocontent of this enzyme was also increased in the cerebral cortex and decreased in the hippocampus. Levels of acetylcholinesterase mRNA transcripts were not altered. Peripherally, homocysteine increased TNF-α, IL-6, and MCP-1 levels in the heart and IL-6 levels in serum. Taken altogether, these findings suggest that homocysteine promotes an inflammatory status that can contribute, at least in part, to neuronal and cardiovascular dysfunctions observed in mild hyperhomocysteinemia.


Subject(s)
Acetylcholinesterase/metabolism , Cerebral Cortex/metabolism , Cytokines/metabolism , Hippocampus/metabolism , Hyperhomocysteinemia/metabolism , Animals , Cerebral Cortex/pathology , Female , Hippocampus/pathology , Inflammation/metabolism , Inflammation/pathology , RNA, Messenger/metabolism , Rats, Wistar
7.
Mol Cell Biochem ; 389(1-2): 229-38, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24378995

ABSTRACT

In the present study we investigated the effects of lung injury on energy metabolism (succinate dehydrogenase, complex II, cytochrome c oxidase, and ATP levels), respiratory mechanics (dynamic and static compliance, elastance and respiratory system resistance) in the lungs of rats, as well as on phospholipids in bronchoalveolar lavage fluid. The protective effect of physical exercise on the alterations caused by lung injury, including lung edema was also evaluated. Wistar rats were submitted to 2 months of physical exercise. After this period the lung injury was induced by intratracheal instillation of lipopolysaccharide. Adult Wistar rats were submitted to 2 months of physical exercise and after this period the lung injury was induced by intratracheal instillation of lipopolysaccharide in dose 100 µg/100 g body weight. The sham group received isotonic saline instillation. Twelve hours after the injury was performed the respiratory mechanical and after the rats were decapitated and samples were collected. The rats subjected to lung injury presented a decrease in activities of the enzymes of the electron transport chain and ATP levels in lung, as well as the formation of pulmonary edema. A decreased lung dynamic and static compliance, as well as an increase in respiratory system resistance, and a decrease in phospholipids content were observed. Physical exercise was able to totally prevent the decrease in succinate dehydrogenase and complex II activities and the formation of pulmonary edema. It also partially prevented the increase in respiratory system resistance, but did not prevent the decrease in dynamic and static compliance, as well as in phospholipids content. These findings suggest that the mitochondrial dysfunction may be one of the important contributors to lung damage and that physical exercise may be beneficial in this pathology, although it did not prevent all changes present in lung injury.


Subject(s)
Energy Metabolism/physiology , Lung Injury/physiopathology , Lung/physiopathology , Physical Conditioning, Animal/physiology , Respiratory Mechanics/physiology , Adenosine Triphosphate/metabolism , Animals , Bronchoalveolar Lavage Fluid , Disease Models, Animal , Electron Transport/drug effects , Electron Transport/physiology , Energy Metabolism/drug effects , Lipopolysaccharides/pharmacology , Lung/drug effects , Lung/metabolism , Lung Injury/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Phospholipids/metabolism , Pulmonary Edema/metabolism , Pulmonary Edema/physiopathology , Rats , Rats, Wistar , Respiratory Mechanics/drug effects
8.
Mol Cell Biochem ; 378(1-2): 91-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23467881

ABSTRACT

Na(+),K(+)-ATPase is a membrane protein which plays a key role in the maintenance of ion homeostasis that is necessary to neuronal excitability, secondary transport and neurotransmitter uptake. Mild hyperhomocysteinemia leads to several clinical manifestations and particularly cerebral diseases; however, little is known about the mechanisms of homocysteine on cerebral Na(+),K(+)-ATPase. In the present study, we investigated the effect of mild hyperhomocysteinemia on the activity, the immunocontent of catalytic subunits (α1, α2, and α3) and the gene expression of this enzyme. We used the experimental model of mild hyperhomocysteinemia that was induced by homocysteine administration (0.03 µmol/g of body weight) twice a day, from the 30th to the 60th postpartum day. Controls received saline in the same volumes. Results showed that mild hyperhomocysteinemia significantly decreased the activity and the immunocontent of the α 1 and α 2 subunits of the Na(+),K(+)-ATPase in cerebral cortex and hippocampus of adult rats. On the other hand, we did not observe any change in levels of Na(+),K(+)-ATPase mRNA transcripts in such cerebral structures of rats after chronic exposure to homocysteine. The present findings support that the homocysteine modulates the Na(+),K(+)-ATPase and this could be associated, at least in part, with the risk to the development of cerebral diseases in individuals with mild hyperhomocysteinemia.


Subject(s)
Cerebral Cortex/enzymology , Hyperhomocysteinemia/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Transcription, Genetic , Animals , Blotting, Western , Catalytic Domain , Hippocampus/enzymology , Homocysteine , Hyperhomocysteinemia/chemically induced , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/genetics
9.
Cell Biochem Funct ; 31(7): 575-84, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23225327

ABSTRACT

Homocystinuria is a neurometabolic disease caused by a severe deficiency of cystathionine beta-synthase activity, resulting in severe hyperhomocysteinemia. Affected patients present several symptoms including a variable degree of motor dysfunction. In this study, we investigated the effect of chronic hyperhomocysteinemia on the cell viability of the mitochondrion, as well as on some parameters of energy metabolism, such as glucose oxidation and activities of pyruvate kinase, citrate synthase, isocitrate dehydrogenase, malate dehydrogenase, respiratory chain complexes and creatine kinase in gastrocnemius rat skeletal muscle. We also evaluated the effect of creatine on biochemical alterations elicited by hyperhomocysteinemia. Wistar rats received daily subcutaneous injections of homocysteine (0.3-0.6 µmol/g body weight) and/or creatine (50 mg/kg body weight) from the 6th to the 28th days of age. The animals were decapitated 12 h after the last injection. Homocysteine decreased the cell viability of the mitochondrion and the activities of pyruvate kinase and creatine kinase. Succinate dehydrogenase was increased other evaluated parameters were not changed by this amino acid. Creatine, when combined with homocysteine, prevented or caused a synergistic effect on some changes provoked by this amino acid. Creatine per se or creatine plus homocysteine altered glucose oxidation. These findings provide insights into the mechanisms by which homocysteine exerts its effects on skeletal muscle function, more studies are needed to elucidate them. Although creatine prevents some alterations caused by homocysteine, it should be used with caution, mainly in healthy individuals because it could change the homeostasis of normal physiological functions.


Subject(s)
Creatine/pharmacology , Homocysteine/metabolism , Hyperhomocysteinemia/metabolism , Muscle, Skeletal/drug effects , Animals , Cell Survival/drug effects , Citric Acid Cycle , Creatine/therapeutic use , Drug Synergism , Energy Metabolism , Female , Glucose/metabolism , Homocysteine/pharmacology , Hyperhomocysteinemia/drug therapy , Hyperhomocysteinemia/pathology , Male , Mitochondria/drug effects , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Oxidation-Reduction , Rats , Rats, Wistar
10.
Exp Cell Res ; 319(3): 89-104, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23142028

ABSTRACT

Hyperprolinemia is an inherited disorder of proline (Pro) metabolism and patients affected by this disease may present neurological manifestations. However, the mechanisms of neural excitotoxicity elicited by hyperprolinemia are far from being understood. Considering the pivotal role of cytoskeletal remodeling in several neurodegenerative pathologies and the potential links between cytoskeleton, reactive oxygen species production and cell death, the aim of the present work was to study the effects of Pro on astrocyte and neuron cytoskeletal remodeling and the possible oxidative stress involvement. Pro induced a shift of actin cytoskeleton in stress fibers together with increased RhoA immunocontent and ERK1/2 phosphorylation/activation in cortical astrocytes. Unlike astrocytes, results evidenced little susceptibility of neuron cytoskeleton remodeling, since Pro-treated neurons presented unaltered neuritogenesis. We observed increased hydrogen peroxide production characterizing oxidative stress together with decreased superoxide dismutase (SOD) and catalase (CAT) activities in cortical astrocytes after Pro treatment, while glutathione peroxidase (GSHPx) activity remained unaltered. However, coincubation with Pro and Trolox/melatonin prevented decreased SOD and CAT activities in Pro-treated astrocytes. Accordingly, these antioxidants were able to prevent the remodeling of the actin cytoskeleton, RhoA increased levels and ERK1/2 phosphorylation in response to high Pro exposure. Taken together, these findings indicated that the cytoskeleton of cortical astrocytes, but not of neurons in culture, is a target to Pro and such effects could be mediated, at least in part, by redox imbalance, RhoA and ERK1/2 signaling pathways. The vulnerability of astrocyte cytoskeleton may have important implications for understanding the effects of Pro in the neurotoxicity linked to inborn errors of Pro metabolism.


Subject(s)
Astrocytes/drug effects , Cerebral Cortex/drug effects , Cytoskeleton/drug effects , Oxidative Stress/drug effects , Proline/pharmacology , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Animals, Newborn , Antioxidants/metabolism , Astrocytes/metabolism , Astrocytes/physiology , Astrocytes/ultrastructure , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , Cytoskeleton/metabolism , Cytoskeleton/physiology , Embryo, Mammalian , Oxidative Stress/physiology , Proline/adverse effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
11.
Metab Brain Dis ; 27(4): 605-12, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22968482

ABSTRACT

The use of psychostimulant methylphenidate has increased in recent years for the treatment of attention-deficit hyperactivity disorder in children and adolescents. However, the behavioral and neurochemical changes promoted by its use are not yet fully understood, particularly when used for a prolonged period during stages of brain development. Thus, the aim of this study was to determine some parameters of oxidative stress in encephalic structures of juvenile rats subjected to chronic methylphenidate treatment. Wistar rats received intraperitoneal injections of methylphenidate (2.0 mg/kg) once a day, from the 15th to the 45th day of age or an equivalent volume of 0.9% saline solution (controls). Two hours after the last injection, animals were euthanized and the encephalic structures obtained for determination of oxidative stress parameters. Results showed that methylphenidate administration increased the activities of superoxide dismutase and catalase, but did not alter the levels of reactive species, thiobarbituric acid reactive substances levels and sulfhydryl group in cerebellum of rats. In striatum and hippocampus, the methylphenidate-treated rats presented a decrease in the levels of reactive species and thiobarbituric acid reactive substances, but did not present changes in the sulfhydryl groups levels. In prefrontal cortex, methylphenidate promoted an increase in reactive species formation, SOD/CAT ratio, and increased the lipid peroxidation and protein damage. These findings suggest that the encephalic structures respond differently to methylphenidate treatment, at least, when administered chronically to young rats. Notably, the prefrontal cortex of juvenile rats showed greater sensitivity to oxidative effects promoted by methylphenidate in relation to other encephalic structures analyzed.


Subject(s)
Central Nervous System Stimulants/toxicity , Methylphenidate/toxicity , Prefrontal Cortex/metabolism , Animals , Antioxidants/metabolism , Catalase/metabolism , Cerebellum/drug effects , Cerebellum/metabolism , Fluoresceins , Glutathione Peroxidase/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Injections, Intraperitoneal , Neostriatum/drug effects , Neostriatum/metabolism , Nitric Oxide/metabolism , Oxidative Stress/physiology , Prefrontal Cortex/pathology , Rats , Rats, Wistar , Reactive Nitrogen Species/metabolism , Sulfhydryl Compounds/metabolism , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
12.
Mol Cell Biochem ; 362(1-2): 187-94, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22045065

ABSTRACT

Since mild hyperhomocysteinemia is a risk factor for cardiovascular and cerebral diseases and extracellular nucleotides/nucleosides, which are controlled by the enzymatic action of ectonucleotidases, can induce an immune response, in the present study, we investigated the effect of chronic mild hyperhomocysteinemia on ectonucleotidase activities and expression in lymphocytes from mesenteric lymph nodes and serum of adult rats. For the chronic chemically induced mild hyperhomocysteinemia, Hcy (0.03 µmol/g of body weight) or saline (control) were administered subcutaneously from the 30th to the 60th day of life. Results showed that homocysteine significantly decreased ATP, ADP, and AMP hydrolysis in lymphocytes of adult rats. E-NTPDases transcriptions were not affected, while the ecto-5'-nucleotidase transcription was significantly decreased in mesenteric lymph nodes of hyperhomocysteinemic rats. ATP, ADP, and AMP hydrolysis were not affected by homocysteine in rat serum. Our findings suggest that Hcy in levels similar to considered risk factor to development of vascular diseases modulates the ectonucleotidases, which could lead to a pro-inflammatory status.


Subject(s)
5'-Nucleotidase/biosynthesis , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Hyperhomocysteinemia/metabolism , Lymphocytes/metabolism , 5'-Nucleotidase/genetics , Animals , Homocysteine/blood , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/pathology , Lymphocytes/immunology , Lymphocytes/pathology , Mesentery , Rats , Rats, Wistar
14.
J Neural Transm (Vienna) ; 119(6): 661-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22143406

ABSTRACT

Schizophrenia is a debilitating mental disorder with a global prevalence of 1% and its etiology remains poorly understood. In the current study we investigated the influence of antipsychotic drugs on the effects of MK-801 administration, which is a drug that mimics biochemical changes observed in schizophrenia, on Na(+), K(+)-ATPase activity and some parameters of oxidative stress in zebrafish brain. Our results showed that MK-801 treatment significantly decreased Na(+), K(+)-ATPase activity, and all antipsychotics tested prevented such effects. Acute MK-801 treatment did not alter reactive oxygen/nitrogen species by 2'7'-dichlorofluorscein (H2DCF) oxidation assay, but increased the levels of thiobarbituric acid reactive substances (TBARS), when compared with controls. Some antipsychotics such as sulpiride, olanzapine, and haloperidol prevented the increase of TBARS caused by MK-801. These findings indicate oxidative damage might be a mechanism involved in the decrease of Na(+), K(+)-ATPase activity induced by MK-801. The parameters evaluated in this study had not yet been tested in this animal model using the MK-801, suggesting that zebrafish is an animal model that can contribute for providing information on potential treatments and disease characteristics.


Subject(s)
Antipsychotic Agents/pharmacology , Brain Chemistry/drug effects , Dizocilpine Maleate/antagonists & inhibitors , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Oxidative Stress/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Zebrafish/metabolism , Animals , Benzodiazepines/pharmacology , Female , Fluoresceins/metabolism , Haloperidol/pharmacology , Lipid Peroxidation/drug effects , Male , Membranes/drug effects , Membranes/metabolism , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Olanzapine , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Sulpiride/pharmacology , Thiobarbituric Acid Reactive Substances/metabolism
15.
Int J Dev Neurosci ; 29(7): 693-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21704148

ABSTRACT

The purpose of this study was to develop a chronic chemically induced model of mild hyperhomocysteinemia in adult rats. We produced levels of Hcy in the blood (30µM), comparable to those considered a risk factor for the development of neurological and cardiovascular diseases, by injecting homocysteine subcutaneously (0.03µmol/g of body weight) twice a day, from the 30th to the 60th postpartum day. Controls received saline in the same volumes. Using this model, we evaluated the effect of chronic administration of homocysteine on redox status in the blood and cerebral cortex of adult rats. Reactive oxygen species and thiobarbituric acid reactive substances were significantly increased in the plasma and cerebral cortex, while nitrite levels were reduced in the cerebral cortex, but not in the plasma, of rats subjected to chronic mild hyperhomocysteinemia. Homocysteine was also seen to disrupt enzymatic and non-enzymatic antioxidant defenses in the blood and cerebral cortex of rats. Since experimental animal models are useful for understanding the pathophysiology of human diseases, the present model of mild hyperhomocysteinemia may be useful for the investigation of additional mechanisms involved in tissue alterations caused by homocysteine.


Subject(s)
Disease Models, Animal , Homocysteine/administration & dosage , Homocysteine/pharmacology , Hyperhomocysteinemia/chemically induced , Oxidative Stress/drug effects , Animals , Antioxidants/metabolism , Catalase/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Glutathione Peroxidase/metabolism , Homocysteine/blood , Humans , Nitrites/metabolism , Oxidation-Reduction , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
16.
Neurobiol Learn Mem ; 94(3): 312-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20659572

ABSTRACT

Since previous studies have shown that ovariectomy impairs memory and cognition, we investigated whether physical exercise would affect ovariectomy-induced memory deficits in inhibitory avoidance and Morris water maze tasks. Female adult Wistar rats were assigned to one of the following groups: sham (submitted to surgery without removal of the ovaries), exercise, ovariectomy (Ovx) and Ovx plus exercise. Thirty days after ovariectomy or sham surgery, animals were submitted to 1 month of treadmill exercise training for 20 min, three times per week. Rats were than tested in inhibitory avoidance and Morris water maze tasks in order to verify ovariectomy effects on aversive and spatial memory performance. Results show that ovariectomized rats were impaired in aversive memory and spatial navigation, both in reference and working memory protocols. Confirming the working hypothesis, ovariectomized rats submitted to exercise had those impairments prevented. These findings support that physical exercise might constitute an important strategy to minimize cognitive deficits found in post-menopausal women.


Subject(s)
Avoidance Learning/physiology , Maze Learning/physiology , Physical Conditioning, Animal/physiology , Space Perception/physiology , Spatial Behavior/physiology , Analysis of Variance , Animals , Female , Ovariectomy , Rats , Rats, Wistar
17.
Neurobiol Learn Mem ; 94(2): 247-53, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20561592

ABSTRACT

Methylphenidate, a psychostimulant that affects both dopaminergic and noradrenergic systems, is one of the most frequently prescribed treatments for attention-deficit hyperactivity disorder. The present study investigated the effects of chronic administration of methylphenidate to juvenile rats on spatial memory, brain-derived neurotrophic factor immunocontent and acetylcholinesterase activity in hippocampus and prefrontal cortex. Rats received intraperitoneal injections of methylphenidate (2.0mg/kg) once a day, from the 15th to the 45th day of age or an equivalent volume of 0.9% saline solution (controls). Twenty-four hours after the last injection, animals were subjected to testing in the Morris water maze. After that, animals were sacrificed and hippocampus and prefrontal cortex were dissected out for determination of brain-derived neurotrophic factor immunocontent and acetylcholinesterase activity. Chronic administration of methylphenidate provoked cognitive impairments on spatial reference and working memory tasks. A reduction on brain-derived neurotrophic factor immunocontent and increased acetylcholinesterase activity in prefrontal cortex, but not in hippocampus, of rats treated with methylphenidate were also observed. These results suggest that the deficit in spatial memory may be associated to decreased brain-derived neurotrophic factor immunocontent and increased acetylcholinesterase in prefrontal cortex of juvenile rats subjected to methylphenidate administration.


Subject(s)
Acetylcholinesterase/drug effects , Brain-Derived Neurotrophic Factor/drug effects , Central Nervous System Stimulants/pharmacology , Hippocampus/drug effects , Maze Learning/drug effects , Methylphenidate/pharmacology , Acetylcholinesterase/metabolism , Age Factors , Analysis of Variance , Animals , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/growth & development , Hippocampus/metabolism , Immunohistochemistry , Male , Maze Learning/physiology , Prefrontal Cortex/drug effects , Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Spatial Behavior/drug effects , Spatial Behavior/physiology , Statistics, Nonparametric
18.
Int J Dev Neurosci ; 28(1): 21-30, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19822200

ABSTRACT

The aim of the present work was to investigate the actions of a chemically induced chronic hyperhomocysteinemia model on intermediate filaments (IFs) of cortical and hippocampal neural cells and explore signaling mechanisms underlying such effects. Results showed that in hyperhomocysteinemic rats the expression of neural IF subunits was affected. In cerebral cortex, glial fibrillary acidic protein (GFAP) expression was donwregulated while in hippocampus high and middle molecular weight neurofilament subunits (NF-H and NF-M, respectively) were up-regulated. Otherwise, the immunocontent of IF proteins was unaltered in cerebral cortex while in hippocampus the immunocontent of cytoskeletal-associated low molecular weight neurofilament (NF-L) and NF-H subunits suggested a stoichiometric ratio consistent with a decreased amount of core filaments enriched in lateral projections. These effects were not accompanied by an alteration in IF phosphorylation. In vitro results showed that 500muM Hcy-induced protein phosphatases 1-, 2A- and 2B-mediated hypophosphorylation of NF subunits and GFAP in hippocampal slices of 17-day-old rats without affecting the cerebral cortex, showing a window of vulnerability of cytoskeleton in developing hippocampus. Ionotropic and metabotropic glutamate receptors were involved in this action, as well as Ca(2+) release from intracellular stores through ryanodine receptors. We propose that the mechanisms observed in the hippocampus of 17-day-old rats could support the neural damage observed in these animals.


Subject(s)
Brain/growth & development , Brain/metabolism , Calcium/metabolism , Glutamic Acid/metabolism , Hyperhomocysteinemia/metabolism , Intermediate Filaments/metabolism , Aging , Animals , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Chronic Disease , Disease Models, Animal , Hippocampus/growth & development , Hippocampus/metabolism , In Vitro Techniques , Intermediate Filament Proteins/metabolism , Phosphorylation , Rats , Rats, Wistar , Receptors, Glutamate/metabolism , Receptors, Metabotropic Glutamate/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism
19.
J Neural Transm (Vienna) ; 116(12): 1681-7, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19756363

ABSTRACT

Methylphenidate is a central nervous system stimulant used for the treatment of attention-deficit hyperactivity disorder. Na(+), K(+)-ATPase is a membrane-bound enzyme necessary to maintain neuronal excitability. Considering that methylphenidate effects on central nervous system metabolism are poorly known and that Na(+), K(+)-ATPase is essential to normal brain function, the purpose of this study was to evaluate the effect of this drug on Na(+), K(+)-ATPase activity in the cerebrum of young and adult rats. For acute administration, a single injection of methylphenidate (1.0, 2.0, or 10.0 mg/Kg) or saline was given to rats on postnatal day 25 or postnatal day 60, in the young and adult groups, respectively. For chronic administration, methylphenidate (1.0, 2.0, or 10.0 mg/Kg) or saline injections were given to young rats starting at postnatal day 25 once daily for 28 days. In adult rats, the same regimen was performed starting at postnatal day 60. Our results showed that acute methylphenidate administration increased Na(+), K(+)-ATPase activity in hippocampus, prefrontal cortex, and striatum of young and adult rats. In young rats, chronic administration of methylphenidate also enhanced Na(+), K(+)-ATPase activity in hippocampus and prefrontal cortex, but not in striatum. When tested in adult rats, Na(+), K(+)-ATPase activity was increased in all cerebral structures studied. The present findings suggest that increased Na(+), K(+)-ATPase activity may be associated with neuronal excitability caused by methylphenidate.


Subject(s)
Central Nervous System Stimulants/pharmacology , Cerebrum/drug effects , Cerebrum/enzymology , Methylphenidate/pharmacology , Sodium-Potassium-Exchanging ATPase/metabolism , Aging , Animals , Central Nervous System Stimulants/administration & dosage , Cerebrum/metabolism , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/metabolism , Male , Methylphenidate/administration & dosage , Prefrontal Cortex/drug effects , Prefrontal Cortex/enzymology , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Time Factors
20.
J Neural Transm (Vienna) ; 116(4): 431-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19255820

ABSTRACT

We evaluated Na(+),K(+)-ATPase activity in hippocampus of rats submitted to an animal model of mania which included the use of lithium and valproate. In the acute treatment, amphetamine or saline was administered to rats for 14 days, between day 8 and 14, rats were treated with lithium, valproate or saline. In the maintenance treatment, rats were treated with lithium, valproate or saline, between day 8 and 14, amphetamine or saline were administered. Locomotor activity was assessed by open field test and Na(+),K(+)-ATPase activity was measured. Our results showed that mood stabilizers reversed and prevented amphetamine-induced behavioral effects. Moreover, amphetamine (acute treatment) increased Na(+),K(+)-ATPase activity, and administration of lithium or valproate reversed this effect. In the maintenance treatment, amphetamine increased Na(+),K(+)-ATPase activity in saline-pretreated rats. Amphetamine administration in lithium- or valproate-pretreated animals did not alter Na(+),K(+)-ATPase activity. The findings suggest that amphetamine-induced hyperactivity may be associated with an increase in Na(+),K(+)-ATPase.


Subject(s)
Bipolar Disorder/enzymology , Hippocampus/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Amphetamine , Analysis of Variance , Animals , Antimanic Agents/therapeutic use , Bipolar Disorder/chemically induced , Bipolar Disorder/drug therapy , Disease Models, Animal , Lithium Compounds/therapeutic use , Male , Motor Activity/drug effects , Rats , Rats, Wistar , Valproic Acid/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...