Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 43
1.
Front Psychiatry ; 12: 701408, 2021.
Article En | MEDLINE | ID: mdl-34421682

Major depressive disorder (MDD) leads to pervasive changes in the health of afflicted patients. Despite advances in the understanding of MDD and its treatment, profound innovation is needed to develop fast-onset antidepressants with higher effectiveness. When acutely administered, the endogenous nucleoside guanosine (GUO) shows fast-onset antidepressant-like effects in several mouse models, including the olfactory bulbectomy (OBX) rodent model. OBX is advocated to possess translational value and be suitable to assess the time course of depressive-like behavior in rodents. This study aimed at investigating the long-term behavioral and neurochemical effects of GUO in a mouse model of depression induced by bilateral bulbectomy (OBX). Mice were submitted to OBX and, after 14 days of recovery, received daily (ip) administration of 7.5 mg/kg GUO or 40 mg/kg imipramine (IMI) for 45 days. GUO and IMI reversed the OBX-induced hyperlocomotion and recognition memory impairment, hippocampal BDNF increase, and redox imbalance (ROS, NO, and GSH levels). GUO also mitigated the OBX-induced hippocampal neuroinflammation (IL-1, IL-6, TNF-α, INF-γ, and IL-10). Brain microPET imaging ([18F]FDG) shows that GUO also prevented the OBX-induced increase in hippocampal FDG metabolism. These results provide additional evidence for GUO antidepressant-like effects, associated with beneficial neurochemical outcomes relevant to counteract depression.

2.
Purinergic Signal ; 17(2): 255-271, 2021 06.
Article En | MEDLINE | ID: mdl-33834349

Ischemic stroke is a major cause of morbidity and mortality worldwide and only few affected patients are able to receive treatment, especially in developing countries. Detailed pathophysiology of brain ischemia has been extensively studied in order to discover new treatments with a broad therapeutic window and that are accessible to patients worldwide. The nucleoside guanosine (Guo) has been shown to have neuroprotective effects in animal models of brain diseases, including ischemic stroke. In a rat model of focal permanent ischemia, systemic administration of Guo was effective only when administered immediately after stroke induction. In contrast, intranasal administration of Guo (In-Guo) was effective even when the first administration was 3 h after stroke induction. In order to validate the neuroprotective effect in this larger time window and to investigate In-Guo neuroprotection under global brain dysfunction induced by ischemia, we used the model of thermocoagulation of pial vessels in Wistar rats. In our study, we have found that In-Guo administered 3 h after stroke was capable of preventing ischemia-induced dysfunction, such as bilateral suppression and synchronicity of brain oscillations and ipsilateral cell death signaling, and increased permeability of the blood-brain barrier. In addition, In-Guo had a long-lasting effect on preventing ischemia-induced motor impairment. Our data reinforce In-Guo administration as a potential new treatment for brain ischemia with a more suitable therapeutic window.


Brain/physiopathology , Guanosine/administration & dosage , Guanosine/therapeutic use , Ischemic Stroke/drug therapy , Ischemic Stroke/physiopathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Administration, Intranasal , Animals , Blood-Brain Barrier/drug effects , Cell Death/drug effects , Cerebral Veins/drug effects , Electrocoagulation , Electroencephalography/drug effects , Functional Laterality/drug effects , Ischemic Stroke/complications , Male , Movement Disorders/etiology , Movement Disorders/prevention & control , Rats , Rats, Wistar , Signal Transduction/drug effects
3.
Neurochem Int ; 124: 114-122, 2019 03.
Article En | MEDLINE | ID: mdl-30639195

OBJECTIVE: Some factors related to lifestyle, including stress and high-fat diet (HFD) consumption, are associated with higher prevalence of obesity. These factors can lead to an imbalance between ROS production and antioxidant defenses and to mitochondrial dysfunctions, which, in turn, could cause metabolic impairments, favoring the development of obesity. However, little is known about the interplay between these factors, particularly at early ages, and whether long-term sex-specific changes may occur. Here, we evaluated whether social isolation during the prepubertal period only, associated or not with chronic HFD, can exert long-term effects on oxidative status parameters and on mitochondrial function in the whole hypothalamus, in a sex-specific manner. METHODS: Wistar male and female rats were divided into two groups (receiving standard chow or standard chow + HFD), that were subdivided into exposed or not to social isolation during the prepubertal period. Oxidative status parameters, and mitochondrial function were evaluated in the hypothalamus in the adult age. RESULTS: Regarding antioxidant enzymes activities, HFD decreased GPx activity in the hypothalamus, while increasing SOD activity in females. Females also presented increased total thiols; however, non-protein thiols were lower. Main effects of stress and HFD were observed in TBARS levels in males, with both factors decreasing this parameter. Additionally, HFD increased complex IV activity, and decreased mitochondrial mass in females. Complex I-III activity was higher in males compared to females. CONCLUSION: Stress during the prepubertal period and chronic consumption of HFD had persistent sex-specific effects on oxidative status, as well as on its consequences for the cell and for mitochondrial function. HFD had more detrimental effects on females, inducing oxidative imbalance, which resulted in damage to the mitochondria. This HFD-induced imbalance may be related to the development of obesity.


Diet, High-Fat/adverse effects , Hypothalamus/metabolism , Mitochondria/metabolism , Oxidative Stress/physiology , Sex Characteristics , Stress, Psychological/metabolism , Animals , Female , Male , Membrane Potentials/physiology , Rats , Rats, Wistar , Sexual Maturation/physiology , Stress, Psychological/psychology
4.
Neurotoxicology ; 67: 305-312, 2018 07.
Article En | MEDLINE | ID: mdl-29680360

The development of new antiepileptic drugs is a high-risk/high-cost research field, which is made even riskier if the behavioral epileptic seizure profile is the unique approach on which the development is based. In order to increase the effectiveness of the screening conducted in the zebrafish model of status epilepticus (SE), the evaluation of neurochemical markers of SE would be of great relevance. Epilepsy is associated with changes in the glutamatergic system, and glutamate uptake is one of the critical parameters of this process. Therefore, we evaluated the levels of glutamate uptake in the zebrafish brain and analyzed its correlation with the progression of behavioral changes in zebrafish at different times after the administration of kainic acid (5 mg/kg). The results showed that the zebrafish suffered with lethargy while swimming for up to 72 h after SE, had reduced levels of GFAP cells 12 h after SE, reduced levels of S100B up to 72 h after SE, and reduced levels of glutamate uptake in the forebrain between 3 h and 12 h after SE. The forebrain region of adult zebrafish after SE present similar changes to the neurochemical limbic alterations that are seen in rodent models of SE. This study demonstrated that there is a time window in which to use the KA zebrafish model of SE to explore some of the known neurochemical alterations that have been observed in rodent models of epilepsy and epileptic human patients.


Glutamic Acid/metabolism , Kainic Acid/toxicity , Locomotion/drug effects , Prosencephalon/metabolism , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Age Factors , Animals , Locomotion/physiology , Male , Prosencephalon/drug effects , Zebrafish
5.
Eur J Nutr ; 57(5): 1913-1924, 2018 Aug.
Article En | MEDLINE | ID: mdl-28567576

PURPOSE: Previously showed that dietary trans fatty acids (TFAs) may cause systemic inflammation and affect the central nervous system (CNS) in Wistar rats by increased levels of cytokines in the cerebrospinal fluid (CSF) and serum (Longhi et al. Eur J Nutr 56(3):1003-1016, 1). Here, we aimed to clarifying the impact of diets with different TFA concentrations on cerebral tissue, focusing on hippocampus and cortex and behavioral performance. METHODS: Wistar rats were fed either a normolipidic or a hyperlipidic diet for 90 days; diets had the same ingredients except for fat compositions, concentrations, and calories. We used lard in the cis fatty acid (CFA) group and PHSO in the TFA group. The intervention groups were as follows: (1) low lard (LL), (2) high lard (HL), (3) low partially hydrogenated soybean oil (LPHSO), and (4) high partially hydrogenated soybean oil (HPHSO). Mitochondrial parameters, tumor necrosis factor alpha (TNF-α), 2'7'-dichlorofluorescein (DCFH) levels in brain tissue, and open field task were analyzed. RESULTS: A worse brain tissue response was associated with oxidative stress in cortex and hippocampus as well as impaired inflammatory and mitochondrial parameters at both PHSO concentrations and there were alterations in the behavioral performance. In many analyses, there were no significant differences between the LPHSO and HPHSO diets. CONCLUSIONS: Partially hydrogenated soybean oil impaired cortical mitochondrial parameters and altered inflammatory and oxidative stress responses, and the hyperlipidic treatment caused locomotor and exploratory effects, but no differences on weight gain in all treatments. These findings suggest that quality is more important than the quantity of fat consumed in terms of CFA and TFA diets.


Dietary Fats/pharmacology , Hippocampus/drug effects , Mitochondria/drug effects , Oxidative Stress/drug effects , Trans Fatty Acids/pharmacology , Animals , Diet , Dietary Fats/administration & dosage , Hippocampus/metabolism , Inflammation/blood , Male , Mitochondria/metabolism , Rats , Rats, Wistar , Soybean Oil , Trans Fatty Acids/administration & dosage
6.
Mol Neurobiol ; 55(2): 980-988, 2018 02.
Article En | MEDLINE | ID: mdl-28084592

In the current study, we verified the effects of maternal hypermethioninemia on the number of neurons, apoptosis, nerve growth factor, and brain-derived neurotrophic factor levels, energy metabolism parameters (succinate dehydrogenase, complex II, and cytochrome c oxidase), expression and immunocontent of Na+,K+-ATPase, edema formation, inflammatory markers (tumor necrosis factor-alpha and interleukin-6), and mitochondrial hydrogen peroxide levels in the encephalon from the offspring. Pregnant Wistar rats were divided into two groups: the first one received saline (control) and the second group received 2.68 µmol methionine/g body weight by subcutaneous injections twice a day during gestation (approximately 21 days). After parturition, pups were killed at the 21st day of life for removal of encephalon. Neuronal staining (anti-NeuN) revealed a reduction in number of neurons, which was associated to decreased nerve growth factor and brain-derived neurotrophic factor levels. Maternal hypermethioninemia also reduced succinate dehydrogenase and complex II activities and increased expression and immunocontent of Na+,K+-ATPase alpha subunits. These results indicate that maternal hypermethioninemia may be a predisposing factor for damage to the brain during the intrauterine life.


Amino Acid Metabolism, Inborn Errors/metabolism , Brain/metabolism , Energy Metabolism/physiology , Glycine N-Methyltransferase/deficiency , Nerve Growth Factors/metabolism , Neurons/metabolism , Prenatal Exposure Delayed Effects/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Amino Acid Metabolism, Inborn Errors/chemically induced , Animals , Cell Count , Female , Glycine N-Methyltransferase/metabolism , Methionine , Oxidation-Reduction , Pregnancy , Rats , Rats, Wistar
7.
Neurochem Res ; 42(11): 3331-3340, 2017 Nov.
Article En | MEDLINE | ID: mdl-28913592

Chronic restraint stress (CRS) induces a variety of changes in brain function, some of which are mediated by glucocorticoids. The response to stress occurs in a sex-specific way, and may include mitochondrial and synaptic alterations. The synapse is highly dependent on mitochondrial energy supply, and when mitochondria become dysfunctional, they orchestrate cell death. This study aimed to investigate the CRS effects on mitochondrial respiratory chain activity, as well as mitochondrial potential and mass in cell body and synapses using hippocampus, cortex and striatum of male and female rats. Rats were divided into non-stressed (control) and stressed group (CRS during 40 days). Results showed that CRS increased complex I-III activity in hippocampus. We also observed an interaction between CRS and sex in the striatal complex II activity, since CRS induced a reduction in complex II activity in males, while in females this activity was increased. Also an interaction was observed between stress and sex in cortical complex IV activity, since CRS induced increased activity in females, while it was reduced in males. Glucocorticoid receptor (GR) content in cortex and hippocampus was sexually dimorphic, with female rats presenting higher levels compared to males. No changes were observed in GR content, mitochondrial potential or mass of animals submitted to CRS. It was concluded that CRS induced changes in respiratory chain complex activities, and some of these changes are sex-dependent: these activities are increased in the striatal mitochondria by CRS protocol mainly in females, while in males it is decreased.


Brain/metabolism , Electron Transport/physiology , Mitochondria/metabolism , Sex Characteristics , Stress, Psychological/metabolism , Animals , Brain/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Chronic Disease , Corpus Striatum/metabolism , Corpus Striatum/pathology , Female , Hippocampus/metabolism , Hippocampus/pathology , Male , Mitochondria/pathology , Rats , Rats, Wistar , Restraint, Physical , Stress, Psychological/pathology
8.
Nutr Res ; 38: 52-63, 2017 Feb.
Article En | MEDLINE | ID: mdl-28381354

Because homocysteine (Hcy) is a risk factor for cardiovascular disease, and vitamin D deficiency can contribute to cardiovascular pathologies. In the present study, we tested the hypothesis that Hcy could impair energy metabolism, mitochondrial function, and redox status in heart slices of Wistar rats and that 1,25-dihydroxivitamin D3 (calcitriol) treatment could prevent such effects. Heart slices were first pretreated with 3 different concentrations of calcitriol (50, 100, and 250nmol/L) for 30minutes at 37°C, after which Hcy was added to promote deleterious effects on metabolism. After 1 hour of incubation, the samples were washed, homogenized, and stored at -80°C before analysis. The results showed that Hcy caused changes in energy metabolism (respiratory chain enzymes), mitochondrial function, and cell viability. Homocysteine also induced oxidative stress, increasing lipid peroxidation, reactive oxygen species generation, and protein damage. An imbalance in antioxidant enzymes was also observed. Calcitriol (50nmol/L) reverted the effect of Hcy on the parameters tested, except for the immunocontent of catalase. Both treatments (calcitriol and Hcy) did not alter the vitamin D receptor immunocontent, which combined with the fact that our ex vivo model is acute, suggesting that the beneficial effect of calcitriol occurs directly through antioxidative mechanisms and not via gene expression. In this study, we show that Hcy impairs mitochondrial function and induces changes in the redox status in heart slices, which were reverted by calcitriol. These findings suggest that calcitriol may be a preventive/therapeutic strategy for complications caused by Hcy.


Antioxidants/pharmacology , Calcitriol/pharmacology , Heart/drug effects , Homocysteine/metabolism , Mitochondria/drug effects , Oxidative Stress/drug effects , Vitamin D/analogs & derivatives , Animals , Antioxidants/metabolism , Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/prevention & control , Cell Survival , Energy Metabolism , Heart/physiopathology , Homocysteine/pharmacology , Lipid Peroxidation , Male , Mitochondria/metabolism , Mitochondria/physiology , Oxidation-Reduction , Protein Carbonylation , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptors, Calcitriol/metabolism , Vitamin D/pharmacology
9.
Article En | MEDLINE | ID: mdl-28223107

Major depressive disorder (MDD) is a neuropsychiatric disease that is associated with profound disturbances in affected individuals. Elucidating the pathophysiology of MDD has been frustratingly slow, especially concerning the neurochemical events and brain regions associated with disease progression. Thus, we evaluated the time-course (up to 8weeks) behavioral and biochemical effects in mice that underwent to a bilateral olfactory bulbectomy (OBX), which is used to modeling depressive-like behavior in rodents. Similar to the symptoms in patients with MDD, OBX induced long-lasting (e.g., impairment of habituation to novelty, hyperactivity and an anxiety-like phenotype) and transient (e.g., loss of self-care and motivational behavior) behavioral effects. Moreover, OBX temporarily impaired hippocampal synaptosomal mitochondria, in a manner that would be associated with hippocampal-related synaptotoxicity. Finally, long-lasting pro-oxidative (i.e., increased levels of reactive oxygen species and nitric oxide and decreased glutathione levels) and pro-inflammatory (i.e., increased levels of pro-inflammatory cytokines IL-1, IL-6, TNF-α and decreased anti-inflammatory cytokine IL-10 levels) effects were induced in the hippocampus by OBX. Additionally, these parameters were transiently affected in the posterior and frontal cortices. This study is the first to suggest that the transient and long-lasting behavioral effects from OBX strongly correlate with mitochondrial, oxidative and inflammatory parameters in the hippocampus; furthermore, these effects show a weak correlation with these parameters in the cortex. Our findings highlight the underlying mechanisms involved in the biochemical time course of events related to depressive behavior.


Behavior, Animal/physiology , Depressive Disorder, Major , Hippocampus , Inflammation , Olfactory Bulb/surgery , Animals , Depressive Disorder, Major/immunology , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Disease Models, Animal , Hippocampus/immunology , Hippocampus/metabolism , Inflammation/immunology , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL
10.
Mol Neurobiol ; 54(6): 4201-4216, 2017 08.
Article En | MEDLINE | ID: mdl-27324900

Although the use, and misuse, of methylphenidate is increasing in childhood and adolescence, there is little information about the consequences of this psychostimulant chronic use on brain and behavior during development. The aim of the present study was to investigate hippocampus biochemical, histochemical, and behavioral effects of chronic methylphenidate treatment to juvenile rats. Wistar rats received intraperitoneal injections of methylphenidate (2.0 mg/kg) or an equivalent volume of 0.9 % saline solution (controls), once a day, from the 15th to the 45th day of age. Results showed that chronic methylphenidate administration caused loss of astrocytes and neurons in the hippocampus of juvenile rats. BDNF and pTrkB immunocontents and NGF levels were decreased, while TNF-α and IL-6 levels, Iba-1 and caspase 3 cleaved immunocontents (microglia marker and active apoptosis marker, respectively) were increased. ERK and PKCaMII signaling pathways, but not Akt and GSK-3ß, were decreased. SNAP-25 was decreased after methylphenidate treatment, while GAP-43 and synaptophysin were not altered. Both exploratory activity and object recognition memory were impaired by methylphenidate. These findings provide additional evidence that early-life exposure to methylphenidate can have complex effects, as well as provide new basis for understanding of the biochemical and behavioral consequences associated with chronic use of methylphenidate during central nervous system development.


Astrocytes/pathology , Behavior, Animal/drug effects , Hippocampus/pathology , Methylphenidate/toxicity , Neurons/pathology , Animals , Antigens, Nuclear/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Cytokines/metabolism , Exploratory Behavior/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glial Fibrillary Acidic Protein/metabolism , Maze Learning/drug effects , Memory/drug effects , Models, Biological , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar , Recognition, Psychology , Signal Transduction , Synaptosomal-Associated Protein 25/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Purinergic Signal ; 12(2): 303-12, 2016 06.
Article En | MEDLINE | ID: mdl-26910734

Glioblastoma multiforme (GBM) is a deadly cancer characterized by a pro-tumoral immune response. T-regulatory (Treg) lymphocytes suppress effector immune cells through cytokine secretion and the adenosinergic system. Ecto-5'-nucleotidase/CD73 plays a crucial role in Treg-mediated immunosuppression in the GBM microenvironment (GME). Methotrexate (MTX) is an immunosuppressive drug that can increase the extracellular concentration of adenosine. In this manuscript, C6 GBM cells were treated with 1.0 µM MTX, and ecto-5'-nucleotidase/CD73 expression and extracellular AMP metabolism were analyzed in vitro. For in vivo studies, rats with implanted GBM were treated for 10 days with MTX-loaded lipid-core nanocapsules (MTX-LNCs, 1 mg/kg/day). The activity of ectonucleotidase and the expression of NTPDase1/CD39 and ecto-5'-nucleotidase/CD73 were measured. The frequencies of T lymphocytes (CD3(+)CD4(+), CD3(+)CD8(+), and CD4(+)CD25(high)CD39(+)) were quantified. In vitro, treatment with MTX increased CD73 expression and activity in C6 cells, which is in agreement with higher levels of extracellular adenosine. In vivo, MTX-LNC treatment increased CD39 expression on CD3(+)CD8(+) lymphocytes. In addition, MTX-LNC treatment up-regulated CD73 expression in tissue isolated from GBM, a finding that is in agreement with the higher activity of this enzyme. More specifically, the treatment increased CD73 expression on CD3(+)CD4(+) and CD3(+)CD8(+) lymphocytes. Treatment with MTX-LNCs decreased the frequencies of T-cytotoxic, T-helper, and Treg lymphocytes in the GME. Although more studies are necessary to better understand the complex cross-talk mediated by supra-physiological concentrations of adenosine in the GME, these studies demonstrate that MTX treatment increases CD73 enzyme expression and AMP hydrolysis, leading to an increase in adenosine production and immunosuppressive capability.


5'-Nucleotidase/biosynthesis , Brain Neoplasms/immunology , Glioblastoma/immunology , Immunosuppressive Agents/pharmacology , Methotrexate/pharmacology , T-Lymphocytes/drug effects , Adenosine Monophosphate/metabolism , Animals , Brain Neoplasms/enzymology , Cell Line, Tumor , Chromatography, High Pressure Liquid , Disease Models, Animal , Glioblastoma/enzymology , Immunohistochemistry , Rats , Tumor Escape/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Up-Regulation
12.
Purinergic Signal ; 12(1): 149-59, 2016 Mar.
Article En | MEDLINE | ID: mdl-26695181

In addition to its intracellular roles, the nucleoside guanosine (GUO) also has extracellular effects that identify it as a putative neuromodulator signaling molecule in the central nervous system. Indeed, GUO can modulate glutamatergic neurotransmission, and it can promote neuroprotective effects in animal models involving glutamate neurotoxicity, which is the case in brain ischemia. In the present study, we aimed to investigate a new in vivo GUO administration route (intranasal, IN) to determine putative improvement of GUO neuroprotective effects against an experimental model of permanent focal cerebral ischemia. Initially, we demonstrated that IN [(3)H] GUO administration reached the brain in a dose-dependent and saturable pattern in as few as 5 min, presenting a higher cerebrospinal GUO level compared with systemic administration. IN GUO treatment started immediately or even 3 h after ischemia onset prevented behavior impairment. The behavior recovery was not correlated to decreased brain infarct volume, but it was correlated to reduced mitochondrial dysfunction in the penumbra area. Therefore, we showed that the IN route is an efficient way to promptly deliver GUO to the CNS and that IN GUO treatment prevented behavioral and brain impairment caused by ischemia in a therapeutically wide time window.


Brain Ischemia/drug therapy , Guanosine/administration & dosage , Guanosine/therapeutic use , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Administration, Intranasal , Animals , Behavior, Animal , Brain Ischemia/psychology , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Guanosine/cerebrospinal fluid , Guanosine/pharmacokinetics , Male , Mitochondria/drug effects , Neuroprotective Agents/cerebrospinal fluid , Neuroprotective Agents/pharmacokinetics , Rats , Rats, Wistar , Stroke/psychology
13.
Int J Dev Neurosci ; 48: 71-9, 2016 Feb.
Article En | MEDLINE | ID: mdl-26658316

Elevated plasma homocysteine (Hcy) levels have been detected in patients with various neurodegenerative conditions. Studies of brain tissue have revealed that hyperhomocysteinemia may impair energy metabolism, resulting in neuronal damage. In addition, new evidence has indicated that vitamin D plays crucial roles in brain development, brain metabolism and neuroprotection. The aim of this study was to investigate the neuroprotective effects of 1,25-dihydroxivitamin D3 (calcitriol) in cerebral cortex slices that were incubated with a mild concentration of Hcy. Cerebral cortex slices from adult rats were first pre-treated for 30 min with one of three different concentrations of calcitriol (50 nM, 100 nM and 250 nM), followed by Hcy for 1h to promote cellular dysfunction. Hcy caused changes in bioenergetics parameters (e.g., respiratory chain enzymes) and mitochondrial functions by inducing changes in mitochondrial mass and swelling. Here, we used flow cytometry to analyze neurons that were double-labelled with Propidium Iodide (PI) and found that Hcy induced an increase in NeuN(+)/PI cells but did not affect GFAP(+)/Pi cells. Hcy also induced oxidative stress by increasing reactive oxygen species generation, lipid peroxidation and protein damage and reducing the activity of antioxidant enzymes (e.g., SOD, CAT and GPx). Calcitriol (50 nM) prevented these alterations by increasing the level of the vitamin D receptor. Our findings suggest that using calcitriol may be a therapeutic strategy for treating the cerebral complications caused by Hcy.


Calcitriol/pharmacology , Cerebral Cortex/drug effects , Homocysteine/pharmacology , Neuroprotective Agents/pharmacology , Animals , Antioxidants/metabolism , Dose-Response Relationship, Drug , Electron Transport Complex II/metabolism , Electron Transport Complex IV/metabolism , Flow Cytometry , Glial Fibrillary Acidic Protein/metabolism , In Vitro Techniques , Male , Phosphopyruvate Hydratase/metabolism , Propidium/metabolism , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/metabolism
14.
Neurochem Res ; 40(9): 1870-81, 2015 Sep.
Article En | MEDLINE | ID: mdl-26232249

Chronic dietary long-chain polyunsaturated fatty acids (PUFAs) deficiency may lead to changes in cortex and hippocampus neuronal membrane phospholipids, and may be linked to impaired central nervous system function. Particularly docosahexaenoic acid deficiency appears to be involved in neuropsychiatric disorders. On the other hand, adverse events early in life may also profoundly affect brain development, leading to long-lasting effects on neurophysiology, neurobiology and behavior. This research assessed if neonatal stress and a dietary n-3 PUFAs deficiency could interact to produce hippocampal alterations related to mitochondrial functions in adult rats. There were no effects of diet, neonatal intervention or interactions on superoxide dismutase or catalase enzymatic activities, mitochondrial membrane potential and respiratory chain complexes. Rats fed n-3 PUFAs deficient diet displayed higher levels of glutathione peroxidase and catalase activity, higher free radicals production and higher thiol content compared to rats fed n-3 PUFAs adequate diet. There were interactions among diets and neonatal stress, since glutathione peroxidase, free radicals production and thiol content were increased in groups that were subjected to neonatal interventions fed n-3 PUFAs deficient diet. Additionally, reduced mitochondrial potential was observed in handled animals. Total thiol revealed a neonatal stress effect, since animals subjected to neonatal interventions displayed lower thiol content. In conclusion, we observed that a chronic treatment with deficient n-3 PUFAs diet, from the puberty period on, increased free radicals production and imbalanced antioxidant enzymes activities, and these increases were higher in animals subjected to neonatal interventions.


Fatty Acids, Omega-3/metabolism , Hippocampus/metabolism , Mitochondria/metabolism , Oxidative Stress , Stress, Physiological , Animals , Animals, Newborn , Female , Membrane Potential, Mitochondrial , Pregnancy , Rats , Rats, Wistar
15.
Biomed Res Int ; 2015: 238010, 2015.
Article En | MEDLINE | ID: mdl-26101767

This study investigated the inhibitory effects of Achyrocline satureioides extract (ASE) incorporated into a topical nanoemulsion on Herpes Simplex Virus type 1 (HSV-1/KOS strain) replication, as well as the distribution of the main ASE flavonoids (quercetin, luteolin, and 3-O-methylquercetin) in porcine skin and mucosa. The ASE-loaded nanoemulsion showed more pronounced effects against HSV-1 replication when compared to the ASE or pure quercetin, as determined by the viral plaque number reduction assay. All flavonoids were detected in the skin epidermis (2.2 µg/cm(2)) and the mucosa upper layers (3.0 µg/cm(2)) from ASE-loaded nanoemulsion until 8 h after topical application. A higher amount of flavonoids was detected when these tissues were impaired, especially in deeper mucosa layers (up to 7-fold). Flavonoids were detected in the receptor fluid only when the mucosa was injured. Such results were supported by confocal microscopy images. Overall, these findings suggest that the tested ASE-loaded nanoemulsion has potential to be used topically for herpes infections.


Emulsions/administration & dosage , Flavonoids/administration & dosage , Mucous Membrane/drug effects , Skin/drug effects , Achyrocline/chemistry , Administration, Topical , Animals , Emulsions/chemistry , Flavonoids/chemistry , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Swine
16.
J Cell Biochem ; 116(10): 2304-12, 2015 Oct.
Article En | MEDLINE | ID: mdl-25833683

The activation of hepatic stellate cell (HSC), from a quiescent cell featuring cytoplasmic lipid droplets to a proliferative myofibroblast, plays an important role in liver fibrosis development. The GRX line is an activated HSC model that can be induced by all-trans-retinol to accumulate lipid droplets. Resveratrol is known for activating Sirtuin1 (SIRT1), a NAD(+)-dependent deacetylase that suppresses the activity of peroxisome proliferator-activated receptor gamma (PPARγ), an important adipogenic transcription factor involved in the quiescence maintenance of HSC. We evaluated the effects of 0.1 µM of resveratrol in retinol-induced GRX quiescence by investigating the interference of SIRT1 and PPARγ on cell lipogenesis. GRX lipid accumulation was evaluated through Oil-red O staining, triacylglycerides quantification, and [(14)C] acetate incorporation into lipids. mRNA expression and protein content of SIRT1 and PPARγ were measured by RT-PCR and immunoblotting, respectively. Resveratrol-mediated SIRT1 stimuli did not induce lipogenesis and reduced the retinol-mediated fat-storing capacity in GRX. In order to support our results, we established a cell culture model of transgenic super expression of PPARγ in GRX cells (GRXPγ). Resveratrol reduced lipid droplets accumulation in GRXPγ cells. These results suggest that the PPARγ/SIRT1 ratio plays an important role in the fate of HSC. Thus, whenever the PPARγ activity is greater than SIRT1 activity the lipogenesis is enabled.


Fibrosis/genetics , Lipid Droplets/metabolism , PPAR gamma/biosynthesis , Sirtuin 1/biosynthesis , Animals , Cell Proliferation , Fibrosis/pathology , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/metabolism , Humans , Kupffer Cells/metabolism , Lipogenesis/drug effects , Liver/metabolism , Mice , Myoblasts/metabolism , PPAR gamma/metabolism , RNA, Messenger/biosynthesis , Resveratrol , Sirtuin 1/metabolism , Stilbenes/administration & dosage , Vitamin A/metabolism
17.
PLoS One ; 10(2): e0116000, 2015.
Article En | MEDLINE | ID: mdl-25706879

BACKGROUND: Step-down inhibitory avoidance task has been widely used to evaluate aversive memory, but crucial parameters inherent to traditional devices that may influence the behavior analysis (as stimulus frequency, animal's bioimpedance) are frequently neglected. NEW METHOD: We developed a new device for step-down inhibitory avoidance task by modifying the shape and distribution of the stainless steel bars in the box floor where the stimuli are applied. The bars are 2 mm wide, with rectangular shape, arranged in pairs at intervals of 1cm from the next pairs. Each pair makes an electrical dipole where the polarity inverts after each pulse. This device also presents a component that acquires and records the exact current received by the animal foot and precisely controls the frequency of stimulus applied during the entire experiment. RESULT: Different from conventional devices, this new apparatus increases the contact surface with bars and animal's paws, allowing the electric current pass through the animal's paws only, drastically reducing the influence of animal's bioimpedance. The analysis of recorded data showed that the current received by the animal was practically the same as applied, independent of the animal's body composition. Importantly, the aversive memory was observed at specific stimuli intensity and frequency (0.35 or 0.5 mA at 62 and 125 Hz but not at 0.20 mA or 20 Hz). Moreover, with this device it was possible to observe the well-known step-down inhibitory avoidance task memory impairment induced by guanosine. CONCLUSION: This new device offers a substantial improvement for behavioral analysis in step-down inhibitory avoidance task and allows us to precisely compare data from different animals with distinct body composition.


Avoidance Learning/physiology , Behavior, Animal/physiology , Body Composition/physiology , Memory/physiology , Animals , Electric Impedance , Electroshock , Rats , Rats, Wistar
18.
Mol Neurobiol ; 52(3): 1791-1803, 2015 Dec.
Article En | MEDLINE | ID: mdl-25394382

Stroke is the major cause of death and the most frequent cause of disability in the adult population worldwide. Guanosine plays an important neuroprotective role in several cerebral ischemic models and is involved in the modulation of oxidative responses and glutamatergic parameters. Because the excessive reactive oxygen species produced during an ischemic event can trigger an inflammatory response, the purpose of this study was to evaluate the hypothesis that guanosine is neuroprotective against focal cerebral ischemia, inhibits microglia/macrophages activation, and mediates an inflammatory response ameliorating the neural damage. Permanent focal cerebral ischemia was induced in adult rats, and guanosine was administered immediately, 1, 3, and 6 h after surgery. Twenty-four hours after ischemia, the asymmetry scores were evaluated by the cylinder test; neuronal damage was evaluated by Fluoro-Jade C (FJC) staining and propidium iodide (PI) incorporation; microglia and immune cells were evaluated by anti-Iba-1 antibody; and inflammatory parameters such as interleukins (IL): IL-1, IL-6, IL-10; tumor necrosis factors alpha (TNF-α); and interferon-gamma (INF-γ) were evaluated in the brain tissue and cerebrospinal fluid. The ischemic event increased the levels of Iba-1-positive cells and pro-inflammatory cytokines and decreased IL-10 levels (an anti-inflammatory cytokine) in the lesion periphery. The guanosine treatment attenuated the changes in these inflammatory parameters and also reduced the infarct volume, PI incorporation, and number of FJC-positive cells, improving the functional recovery. Thus, guanosine may have been a promising therapeutic agent for the treatment of ischemic brain injury by reduction of inflammatory process triggered in an ischemic event.


Anti-Inflammatory Agents/pharmacology , Brain Ischemia/drug therapy , Guanosine/pharmacology , Inflammation/drug therapy , Neuroprotective Agents/pharmacology , Animals , Brain/drug effects , Brain Ischemia/pathology , Cytokines/metabolism , Disease Models, Animal , Male , Microglia/drug effects , Microglia/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism
19.
Cell Biochem Biophys ; 68(2): 247-57, 2014 Mar.
Article En | MEDLINE | ID: mdl-23793960

Resveratrol (RSV) is known for its antioxidant properties; however, this compound has been proposed to have cytotoxic and pro-oxidant effects depending on its concentration and time of exposure. We previously reported the cell cycle arrest effect of low doses of RSV in GRX cells, an activated hepatic stellate cell model. Here, we evaluated the effects of RSV treatment (0.1-50 µM) for 24 and 120 h on GRX viability and oxidative status. Only treatment with 50 µM of RSV reduced the amount of live cells. However, even low doses of RSV induced an increased reactive species production at both treatment times. While being diminished within 24 h, RSV induced an increase in the SOD activity in 120 h. The cellular damage was substantially increased at 24 h in the 50 µM RSV-treated group, as indicated by the high lipoperoxidation, which may be related to the significant cell death and low proliferation. Paradoxically, this cellular damage and lipoperoxidation were considerably reduced in this group after 120 h of treatment while the surviving cells proliferated. In conclusion, RSV induced a dose-dependent pro-oxidant effect in GRX cells. The highest RSV dose induced oxidative-related damage, drastically reducing cell viability; but this cytotoxicity seems to be attenuated during 120 h of treatment.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Hepatic Stellate Cells/drug effects , Stilbenes/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Catalase/metabolism , Cell Line , Cell Survival/drug effects , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Lipid Peroxidation/drug effects , Mice , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Resveratrol , Stilbenes/chemistry , Superoxide Dismutase/metabolism , Time Factors
20.
Neurochem Res ; 38(9): 1791-800, 2013 Sep.
Article En | MEDLINE | ID: mdl-23729300

Social isolation during early development is one of the most potent stressors that can cause alterations in the processes of brain maturation, leading to behavioral and neurochemical changes that may persist to adulthood. Exposure to palatable diets during development can also affect neural circuits with long-term consequences. The aims of the present study were to investigate the long-term effects of isolation stress during the pre-pubertal period on the exploratory and anxiety-like behavior, the oxidative stress parameters and the respiratory chain enzymes activities in the hippocampus of adult male rats under chronic palatable diets. The results showed that isolated rats receiving either normal or high-fat diet during the pre-pubertal period presented an anxiolytic-like behavior. The animals exposed to stress and treated with high-carbohydrate diet, rich in disaccharides, on the other hand, presented the opposite pattern of behavior. Stress in the pre-pubertal period also leads to decreased activity of the antioxidant enzymes and the mitochondrial respiratory chain complexes II and IV and decreased total thiol content. These effects were reversed by high-fat diet when it was associated with stress. The effects of a sub-acute pre-pubertal isolation stress on anxiety-like behavior and on hippocampal oxidative imbalance during adulthood appear to be modulated by different types of diets, and probably different mechanisms are involved.


Anxiety , Behavior, Animal , Diet , Oxidative Stress , Sexual Maturation , Animals , Electron Transport , Male , Rats , Social Isolation
...