Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
Metab Brain Dis ; 38(4): 1155-1166, 2023 04.
Article in English | MEDLINE | ID: mdl-36689104

ABSTRACT

Vitamin D3 deficiency is associated with an increased risk of dementia. An association between vitamin D3 deficiency and subjective cognitive complaints in geriatric patients has been previously reported. This study aimed to evaluate the effects of two doses of vitamin D3 on spatial memory (using the Radial Maze) and cytokine levels [tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6), and interleukin-10 (IL-10)] on 2-, 6-, 13-, 22-, and 31-month-old male Wistar rats. Animals were supplemented with vitamin D3 at doses of 42 IU/kg and 420 IU/kg for 21 days. A radial maze test was performed to evaluate spatial memory. After the behavioral test, the frontal cortex and hippocampus were dissected for enzyme immunoassay analyses to measure the cytokine levels (TNFα, IL-1ß, IL-6, and IL-10). Our results showed that vitamin D3 supplementation reversed spatial memory impairment at the supplemented doses (42 and 420 IU/kg) in 6-, 13-, and 22-month-old animals and at a dose of 420 IU/kg in 31-month-old animals. The lower dose (42 IU/kg) regulates both pro- and anti-inflammatory cytokines mainly in the frontal cortex. Our results suggest that vitamin D3 has a modulatory action on pro- and anti-inflammatory cytokines, since older animals showed increased cytokine levels compared to 2-month-old animals, and that vitamin D3 may exert an immunomodulatory effect on aging.


Subject(s)
Cholecalciferol , Vitamin D Deficiency , Rats , Male , Animals , Cholecalciferol/pharmacology , Cholecalciferol/therapeutic use , Cytokines , Interleukin-10 , Rats, Wistar , Interleukin-6 , Spatial Memory , Tumor Necrosis Factor-alpha , Anti-Inflammatory Agents
2.
Exp Gerontol ; 166: 111873, 2022 09.
Article in English | MEDLINE | ID: mdl-35760268

ABSTRACT

INTRODUCTION: The consumption of soft drinks has increased considerably in recent decades, mainly cola soft drinks. Excessive consumption of cola-based soft drinks is associated with several diseases and cognitive decline, particularly memory impairment. Furthermore, diets with high sugar can promote insulin resistance, metabolic syndrome, and dyslipidemia. AIM: Thus, the present study aimed to evaluate the effect of cola soft drink intake on behavioral alterations and oxidative damage in 2-, 8- and 14- month-old male Wistar rats. METHODS: The soft drink groups drank soft drink and/or water ad libitum during 67 days, the control groups ingested only water. Radial-arm maze and Y-maze were used to evaluate spatial memory, open-field to evaluate the habituation memory, and inhibitory avoidance to evaluate aversive memory. The behavioral tests started at the day 57 and finished at day 67 of treatment. At 68th day, the rats were killed; frontal cortex and hippocampus were dissected to the analysis of antioxidants enzymes catalase (CAT) and superoxide dismutase (SOD); and the oxidative markers thiobarbituric acid reactive substances (TBARS) and dichloro-dihydro-fluorescein diacetate (DCFH) were measured in the hippocampus. RESULTS AND DISCUSSION: The cola-based soft drink intake caused memory impairment in the radial-arm maze, Y-maze task, and open-field in the 2- and 8-month-old rat, but not in the 14-month-old. There were no difference among groups in the inhibitory avoidance test. In the frontal cortex, soft drink intake reduced CAT activity in the 8-month-old rats and SOD activity in the 8- and 14-month-old rats. In the hippocampus, the soft drink increased CAT activity in 2- and 8-month-old rats, increased DCFH levels at all ages, and increased TBARS levels in 2-month-rats. Therefore, the results show that long-term soft drink intake leads to memory impairment and oxidative stress. The younger seems to be more susceptible to the soft drink alterations on behavior; however, soft drink caused alterations in the oxidative system at all ages evaluated.


Subject(s)
Memory Disorders , Oxidative Stress , Animals , Antioxidants/pharmacology , Carbonated Beverages/adverse effects , Hippocampus/metabolism , Male , Maze Learning , Memory Disorders/chemically induced , Memory Disorders/metabolism , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Water/metabolism , Water/pharmacology
3.
Neurochem Res ; 47(3): 613-621, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34674138

ABSTRACT

Critical illness encompasses a wide spectrum of life-threatening clinical conditions requiring intensive care. Our objective was to evaluate cognitive, inflammatory and cellular metabolism alterations in the central nervous system in an animal model of critical illness induced by zymosan. For this Wistar rats that were divided into Sham and zymosan. Zymozan was administered once intraperitoneally (30 g/100 g body weight) diluted in mineral oil. The animals were submitted to behavioral tests of octagonal maze, inhibitory avoidance and elevated plus maze. Brain structures (cortex, prefrontal and hippocampus) were removed at 24 h, 4, 7 and 15 days after zymosan administration for analysis of cytokine levels (TNF-α, IL-1b, IL-6 and IL-10), oxidative damage and oxygen consumption. Zymosan-treated animals presented mild cognitive impairment both in aversive (inhibitory avoidance) and non-aversive (octagonal maze) tasks by day 15. However, they did not show increase in anxiety (elevated-plus maze). The first neurochemical alteration found was an increase in brain pro-inflammatory cytokines (IL-1ß, IL-6 and TNF-α) at day 4th in the hippocampus. In cortex, a late (7 and 15 days) increase in TNF-α was also noted, while the anti-inflammatory cytokine IL-10 decrease from 4 to 15 days. Oxygen consumption was decreased in the hippocampus and pre-frontal, but not cortex, only at 7 days. Additionally, it was observed a late (15 days) increase in oxidative damage parameters. This characterization of brain dysfunction in rodent model of critical illness reproduces some of the alterations reported in humans such neuropsychiatric disorders, especially depression, memory loss and cognitive changes and can add to the nowadays used models.


Subject(s)
Cognitive Dysfunction , Critical Illness , Animals , Brain/metabolism , Cognitive Dysfunction/metabolism , Disease Models, Animal , Hippocampus/metabolism , Oxidative Stress/physiology , Rats , Rats, Wistar , Rodentia
4.
Metab Brain Dis ; 36(8): 2223-2233, 2021 12.
Article in English | MEDLINE | ID: mdl-33950381

ABSTRACT

Although numerous studies have investigated the mechanisms underlying the fast and sustained antidepressant-like effects of ketamine, the contribution of the glucocorticoid receptor (GR) and dendritic branching remodeling to its responses remain to be fully established. This study investigated the ability of a single administration of ketamine to modulate the GR and dendritic branching remodeling and complexity in the hippocampus of mice subjected to chronic corticosterone (CORT) administration. CORT was administered for 21 days, followed by a single administration of ketamine (1 mg ∕kg, i.p.) or fluoxetine (10 mg ∕kg, p.o., conventional antidepressant) in mice. On 22nd, 24 h after the treatments, GR immunocontent in the hippocampus was analyzed by western blotting, while the dendritic arborization and dendrite length in the ventral and dorsal dentate gyrus (DG) of the hippocampus was analyzed by Sholl analysis. Chronic CORT administration downregulated hippocampal GR immunocontent, but this alteration was completely reversed by a single administration of ketamine, but not fluoxetine. Moreover, CORT administration significantly decreased dendritic branching in the dorsal and ventral DG areas and caused a mild decrease in dendrite length in both regions. Ketamine, but not fluoxetine, reversed CORT-induced dendritic branching loss in the ventral and dorsal DG areas, regions associated with mood regulation and cognitive functions, respectively. This study provides novel evidence that a single administration of ketamine, but not fluoxetine, rescued the impairments on GR and dendritic branching in the hippocampus of mice subjected to chronic CORT administration, effects that may be associated with its rapid antidepressant response.


Subject(s)
Ketamine , Animals , Corticosterone/pharmacology , Depression/chemically induced , Fluoxetine/pharmacology , Hippocampus/metabolism , Ketamine/pharmacology , Mice , Receptors, Glucocorticoid
5.
Chem Biol Interact ; 342: 109476, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-33872575

ABSTRACT

Ketamine is the prototype for glutamate-based fast-acting antidepressants. The establishment of ketamine-like drugs is still a challenge and ascorbic acid has emerged as a candidate. This study investigated the ascorbic acid's ability to induce a fast antidepressant-like response and to improve hippocampal synaptic markers in mice subjected to chronic corticosterone (CORT) administration. CORT was administered for 21 days, followed by a single administration of ascorbic acid (1 mg ∕Kg, p.o.), ketamine (1 mg ∕Kg, i.p.) or fluoxetine (10 mg ∕Kg, p.o.) in mice. Depressive-like behavior, hippocampal synaptic proteins immunocontent, dendrite spines density in the dentate gyrus (DG) were analyzed 24 h following treatments. The administration of ascorbic acid or ketamine, but not fluoxetine, counteracted CORT-induced depressive-like behavior in the tail suspension test (TST). CORT administration reduced PSD-95, GluA1, and synapsin (synaptic markers) immunocontent, and these alterations were reversed by ascorbic acid or ketamine, but only ketamine reversed the CORT-induced reduction on GluA1 immunocontent. In the ventral and dorsal DG, CORT decreased filopodia-, thin- and stubby-shaped spines, while ascorbic acid and ketamine abolished this alteration only in filopodia spines. Ascorbic acid and ketamine increased mushroom-shaped spines density in ventral and dorsal DG. Therefore, the results show that a single administration of ascorbic acid, in a way similar to ketamine, rapidly elicits an antidepressant-like response and reverses hippocampal synaptic deficits caused by CORT, an effect associated with increased levels of synaptic proteins and dendritic remodeling.


Subject(s)
Antidepressive Agents/therapeutic use , Ascorbic Acid/therapeutic use , Depression/drug therapy , Hippocampus/drug effects , Animals , Corticosterone , Dendritic Spines/drug effects , Depression/chemically induced , Female , Hindlimb Suspension , Ketamine/therapeutic use , Mice , Neuroprotective Agents/therapeutic use
6.
Metab Brain Dis ; 36(5): 1057-1067, 2021 06.
Article in English | MEDLINE | ID: mdl-33616841

ABSTRACT

D-galactose (D-gal) is a carbohydrate widely distributed in regular diets. However, D-gal administration in rodents is associated with behavioral and neurochemical alterations similar to features observed in aging. In this regard, this study aimed to investigate the effects of D-gal exposure, in different periods, in rats' brain regions' activities of creatine kinase (CK) and tricarboxylic acid (TCA) cycle enzymes. Male adult Wistar rats received D-gal (100 mg/kg, gavage) for 1, 2, 4, 6 or 8 weeks. CK and TCA enzymes' activities were evaluated in rats' prefrontal cortex and hippocampus. In general, the results showed an increase in citrate synthase (CS) and succinate dehydrogenase (SDH) activities in animals treated with D-gal compared to the control group in the prefrontal cortex and hippocampus. Also, in the fourth week, the malate dehydrogenase (MD) activity increased in the hippocampus of rats that received D-gal compared to control rats. In addition, we observed an increase in the CK activity in the prefrontal cortex and hippocampus in the first and eighth weeks of treatment in the D-gal group compared to the control group. D-gal administration orally administered modulated TCA cycle enzymes and CK activities in the prefrontal cortex and hippocampus, which were also observed in aging and neurodegenerative diseases. However, more studies using experimental models are necessary to understand better the impact and contribution of these brain metabolic abnormalities associated with D-gal consumption for aging.


Subject(s)
Brain/drug effects , Citric Acid Cycle/drug effects , Creatine Kinase/metabolism , Galactose/administration & dosage , Malate Dehydrogenase/metabolism , Tricarboxylic Acids/metabolism , Administration, Oral , Animals , Brain/metabolism , Male , Rats , Rats, Wistar
7.
J Gerontol A Biol Sci Med Sci ; 76(6): 991-995, 2021 05 22.
Article in English | MEDLINE | ID: mdl-33249457

ABSTRACT

Folic acid (FA) supplementation is important during pregnancy to avoid malformations in the offspring. However, it is unknown if it can affect the offspring throughout their lives. To evaluate the offspring, female mother rats (dams) were separated into 5 groups: Four groups received the AIN-93 diet, divided into control and FA (5, 10, and 50 mg/kg), and an additional group received a FA-deficient diet, and the diet was performed during pregnancy and lactation. We evaluated the female offspring of these dams (at 2 and 18 months old). The aged offspring fed with FA-deficient diet presented habituation, spatial and aversive memory impairment and the FA maternal supplementation prevented this. The natural aging caused an increase in the TNF-α and IL-1ß levels in the hippocampus from 18-month-old offspring. FA maternal supplementation was able to prevent the increase of these cytokines. IL-4 levels decreased in the prefrontal cortex from aged control rats and FA prevented it. FA deficiency decreased the levels of IL-4 in the hippocampus of the young offspring. In addition, natural aging and FA deficiency decreased brain-derived neurotrophic factor levels in the hippocampus and nerve growth factor levels in the prefrontal cortex and FA supplementation prevented it. Thus, the present study shows for the first time the effect of FA maternal supplementation on memory, cytokines, and neurotrophins in the aged offspring.


Subject(s)
Dietary Supplements , Folic Acid/therapeutic use , Inflammation/prevention & control , Memory Disorders/prevention & control , Prenatal Exposure Delayed Effects/drug therapy , Aging/drug effects , Animals , Female , Folic Acid Deficiency/complications , Hippocampus/metabolism , Inflammation/etiology , Memory Disorders/etiology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Wistar
8.
Metab Brain Dis ; 36(2): 213-224, 2021 02.
Article in English | MEDLINE | ID: mdl-33219893

ABSTRACT

The present study aimed to evaluate the effect of folic acid treatment in an animal model of aging induced by D-galactose (D-gal). For this propose, adult male Wistar rats received D-gal intraperitoneally (100 mg/kg) and/or folic acid orally (5 mg/kg, 10 mg/kg or 50 mg/kg) for 8 weeks. D-gal caused habituation memory impairment, and folic acid (10 mg/kg and 50 mg/kg) reversed this effect. However, folic acid 50 mg/kg per se caused habituation memory impairment. D-gal increased the lipid peroxidation and oxidative damage to proteins in the prefrontal cortex and hippocampus from rats. Folic acid (5 mg/kg, 10 mg/kg, or 50 mg/kg) partially reversed the oxidative damage to lipids in the hippocampus, but not in the prefrontal cortex, and reversed protein oxidative damage in the prefrontal cortex and hippocampus. D-gal induced synaptophysin and BCL-2 decrease in the hippocampus and phosphorylated tau increase in the prefrontal cortex. Folic acid was able to reverse these D-gal-related alterations in the protein content. The present study shows folic acid supplementation as an alternative during the aging to prevent cognitive impairment and brain alterations that can cause neurodegenerative diseases. However, additional studies are necessary to elucidate the effect of folic acid in aging.


Subject(s)
Aging/metabolism , Folic Acid/pharmacology , Habituation, Psychophysiologic/drug effects , Memory Disorders/prevention & control , Oxidative Stress/drug effects , Animals , Galactose , Hippocampus/drug effects , Hippocampus/metabolism , Male , Memory/drug effects , Memory Disorders/chemically induced , Memory Disorders/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar
9.
Mol Neurobiol ; 56(4): 2606-2617, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30051350

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease which is characterized by progressive memory loss, the accumulation of ß-amyloid peptide (Aß) (mainly Aß1-42), and more recently, by neuroinflammation, which has been highlighted as playing a central role in the development and progress of AD. This study utilized 100-day-old Balb/c mice for the induction of an AD-like dementia model. The animals were administered with Aß1-42 oligomers (400 pmol/site) or artificial cerebrospinal fluid (ACSF) into the left cerebral ventricle. Twenty-four hours after intracerebroventricular administration, the animals were treated with minocycline (50 mg/kg, via oral gavage) for 17 days. The animals' locomotion was evaluated using the open-field test. The spatial memory was tested using the Y-maze, and the aversive memory was evaluated using the inhibitory avoidance task. Treatment with minocycline was shown to improve both spatial and aversive memories in mice that were submitted to the dementia model. In addition, minocycline reduced the levels of Aß and microglial activation in the animals that received the administration of Aß1-42 oligomers. Moreover, the results suggest that the decrease in microglial activation occurred because of a reduction in the levels of toll-like receptors 2 (TLR2) content, and its adapter protein MyD88, as well as a reduction in the levels of the protein NLRP3, which is indispensable in the assembly of inflammasome. These observations were evaluated via immunohistochemistry and confirmed using the Western blot analysis. Treatment with minocycline had no effect in preventing apoptotic morphologic alterations of the neurons. Thus, the anti-inflammatory effect of minocycline involves TLR2 receptors and NLRP3, besides being beneficial by ameliorating memory impairments. Graphical Abstract.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/toxicity , Minocycline/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Multimerization , Animals , Cell Survival/drug effects , Locomotion/drug effects , Male , Memory/drug effects , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Toll-Like Receptor 2/metabolism
10.
Exp Gerontol ; 113: 209-217, 2018 11.
Article in English | MEDLINE | ID: mdl-30304709

ABSTRACT

Aging is a complex biological process. Epigenetic alterations have been related to both aging and memory decline. Included amongst these alterations is histone acetylation, which may play a crucial role in aging. Thus, the aims of the present study were to standardize the animal model of d-galactose (d-gal), and to evaluate the effects caused by sodium butyrate (SB), which is a histone deacetylase inhibitor on memory, the modulation of histone deacetylases (HDACs), and also DNA damage in 2, 6 or 16-month-old Wistar rats which were subjected to administrations of d-gal. To help choose the best dose of d-gal for the induction of the aging model, we performed a dose-response curve (100, 200 or 300 mg/kg). d-Gal was administered orally to the 2-month-old rats for a period of 30 days. After this, d-gal (200 mg/kg) or water were administered to the 2, 6 or 16-month-old rats for a period of 30 days. On the 24th day, treatment was started with SB (600 mg/kg) intraperitoneally, for a period of 7 days. SB was able to reverse the damage to habituation memory caused by d-gal in the 2 and 6-month-old rats, but was unable to reverse the damage in the 16 month-old animals. In addition, SB was able to reverse the damage caused by natural aging in the 16-month-old animals. In the inhibitory avoidance task, SB improved the damage caused by d-gal in the 2, 6 and 16-month-old animals and had the same result against the effects of natural aging in the 16-month-old rats. Moreover, d-gal caused an increase in the level of HDACs activity in the 16-month-old animals, and SB was able to reverse this effect in the frontal cortex and hippocampus. The 16-month-old animals showed an increase in the frequency of DNA damage in peripheral blood, and SB was able to reduce this damage. Moreover, d-gal caused an increase in the index and frequency of DNA damage in the 2 and 6-month-old animals, and treatment with SB was able to prevent this damage. Thus, the present study showed the protective effects of SB on the memory of naturally aged and d-gal induced aging in rats. Therefore, the present study shows new findings for the use of SB in aging.


Subject(s)
Aging/drug effects , Butyric Acid/pharmacology , Galactose/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Memory/drug effects , Animals , Brain/drug effects , DNA Damage/drug effects , Disease Models, Animal , Male , Maze Learning/drug effects , Rats , Rats, Wistar
11.
Neurochem Int ; 108: 436-447, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28606823

ABSTRACT

Sepsis is a complication of an infection which imbalance the normal regulation of several organ systems, including the central nervous system (CNS). Evidence points towards inflammation and oxidative stress as major steps associated with brain dysfunction in sepsis. Thus, we investigated the α-lipoic acid (ALA) effect as an important antioxidant compound on brain dysfunction in rats. Wistar rats were subjected to sepsis by cecal ligation and perforation (CLP) or sham (control) and treated orally with ALA (200 mg/kg after CLP) or vehicle. Animals were divided into sham + saline, sham + ALA, CLP + saline and CLP + ALA groups. Twelve, 24 h and 10 days after surgery, the hippocampus, prefrontal cortex and cortex were obtained and assayed for levels of TNF-α and IL-1ß, blood brain barrier (BBB) permeability, nitrite/nitrate concentration, myeloperoxidase (MPO) activity, thiobarbituric acid reactive species (TBARS) formation, protein carbonyls, superoxide dismutase (SOD) and catalase (CAT) activity and neurotrophins levels. Behavioral tasks were performed 10 days after surgery. ALA reduced BBB permeability and TNF-α levels in hippocampus in 24 h and IL-1ß levels and MPO activity in hippocampus and prefrontal cortex in 24 h. ALA reduced nitrite/nitrate concentration and lipid peroxidation in 24 h in all structures and protein carbonylation in 12 and 24 h in hippocampus and cortex. CAT activity increased in the hippocampus and cortex in all times. ALA enhanced NGF levels in hippocampus and cortex and prevented cognitive impairment. Our data demonstrates that ALA reduces the consequences of polymicrobial sepsis in rats by decreasing inflammatory and oxidative stress parameters in the brain.


Subject(s)
Antioxidants/therapeutic use , Cognitive Dysfunction/drug therapy , Coinfection/drug therapy , Inflammation Mediators/antagonists & inhibitors , Sepsis/drug therapy , Thioctic Acid/therapeutic use , Acute Disease , Animals , Antioxidants/pharmacology , Brain/drug effects , Brain/metabolism , Cognitive Dysfunction/metabolism , Coinfection/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Inflammation Mediators/metabolism , Male , Random Allocation , Rats , Rats, Wistar , Sepsis/metabolism , Thioctic Acid/pharmacology , Time Factors
12.
Metab Brain Dis ; 32(3): 811-817, 2017 06.
Article in English | MEDLINE | ID: mdl-28236040

ABSTRACT

D-Galactose (D-gal) chronic administration via intraperitoneal and subcutaneous routes has been used as a model of aging and Alzheimer disease in rodents. Intraperitoneal and subcutaneous administration of D-gal causes memory impairments, a reduction in the neurogenesis of adult mice, an increase in the levels of the amyloid precursor protein and oxidative damage; However, the effects of oral D-gal remain unclear. The aim of this study was to evaluate whether the oral administration of D-gal induces abnormalities within the mitochondrial respiratory chain of rats. Male Wistar rats (4 months old) received D-gal (100 mg/kg v.o.), during the 1st, 2nd, 4th, 6th or 8th weeks by oral gavage. The activity of the mitochondrial respiratory chain complexes was measured in the 1st, 2nd, 4th, 6th and 8th weeks after the administration of D-gal. The activity of the respiratory chain complex I was found to have increased in the prefrontal cortex and hippocampus in the 1st, 6th and 8th weeks, while the activity of the respiratory chain complex II increased in the 1st, 2nd, 4th, 6th and 8th weeks within the hippocampus and in the 2nd, 4th, 6th and 8th weeks within the prefrontal cortex. The activity of complex II-III increased within the prefrontal cortex and hippocampus in each week of oral D-gal treatment. The activity of complex IV increased within the prefrontal cortex and hippocampus in the 1st, 2nd, 6th and 8th weeks of treatment. After 4 weeks of treatment the activity increased only in hippocampus. In conclusion, the present study showed that the oral administration of D-gal increased the activity of the mitochondrial respiratory chain complexes I, II, II-III and IV in the prefrontal cortex and hippocampus. Furthermore, the administration of D-gal via the oral route seems to cause the alterations in the mitochondrial respiratory complexes observed in brain neurodegeneration.


Subject(s)
Electron Transport Complex I/metabolism , Galactose/administration & dosage , Hippocampus/metabolism , Mitochondria/metabolism , Prefrontal Cortex/metabolism , Administration, Oral , Animals , Brain/drug effects , Brain/metabolism , Galactose/toxicity , Hippocampus/drug effects , Male , Mitochondria/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
13.
Mol Neurobiol ; 53(1): 402-407, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25465243

ABSTRACT

Congenital muscular dystrophies 1D (CMD1D) present a mutation on the LARGE gene and are characterized by an abnormal glycosylation of α-dystroglycan (α-DG), strongly implicated as having a causative role in the development of central nervous system abnormalities such as cognitive impairment seen in patients. However, in the animal model of CMD1D, the brain involvement remains unclear. Therefore, the objective of this study is to evaluate the cognitive involvement in the Large(myd) mice. To this aim, we used adult homozygous, heterozygous, and wild-type mice. The mice underwent six behavioral tasks: habituation to an open field, step-down inhibitory avoidance, continuous multiple trials step-down inhibitory avoidance task, object recognition, elevated plus-maze, and forced swimming test. It was observed that Large(myd) individuals presented deficits on the habituation to the open field, step down inhibitory avoidance, continuous multiple-trials step-down inhibitory avoidance, object recognition, and forced swimming. This study shows the first evidence that abnormal glycosylation of α-DG may be affecting memory storage and restoring process in an animal model of CMD1D.


Subject(s)
Behavior, Animal , Muscular Dystrophy, Animal/congenital , Muscular Dystrophy, Animal/pathology , Animals , Avoidance Learning , Disease Models, Animal , Habituation, Psychophysiologic , Maze Learning , Mice, Knockout , Muscular Dystrophy, Animal/physiopathology , Swimming
14.
Muscle Nerve ; 53(1): 91-5, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25965940

ABSTRACT

INTRODUCTION: Duchenne muscular dystrophy (DMD) is a degenerative disease of skeletal, respiratory, and cardiac muscles caused by defects in the dystrophin gene. More recently, brain involvement has been verified. Mitochondrial dysfunction and oxidative stress may underlie the pathophysiology of DMD. In this study we evaluate Krebs cycle enzymes activity in the cerebral cortex, diaphragm, and quadriceps muscles of mdx mice. METHODS: Cortex, diaphragm, and quadriceps tissues from male dystrophic mdx and control mice were used. RESULTS: We observed increased malate dehydrogenase activity in the cortex; increased malate dehydrogenase and succinate dehydrogenase activities in the diaphragm; and increased citrate synthase, isocitrate dehydrogenase, and malate dehydrogenase activities in the quadriceps of mdx mice. CONCLUSION: This study showed increased activity of Krebs cycle enzymes in cortex, quadriceps, and diaphragm in mdx mice.


Subject(s)
Citrate (si)-Synthase/metabolism , Citric Acid Cycle/physiology , Isocitrate Dehydrogenase/metabolism , Malate Dehydrogenase/metabolism , Muscular Dystrophy, Duchenne/enzymology , Animals , Cerebral Cortex/enzymology , Diaphragm/enzymology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/enzymology , Muscular Dystrophy, Duchenne/genetics
15.
Aging Dis ; 6(5): 331-41, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26425388

ABSTRACT

Aging is a normal physiological process accompanied by cognitive decline. This aging process has been the primary risk factor for development of aging-related diseases such as Alzheimer's disease (AD). Cognitive deficit is related to alterations of neurotrophic factors level such as brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF) and glial cell-derived neurotrophic factor (GDNF). These strong relationship between aging and AD is important to investigate the time which they overlap, as well as, the pathophysiological mechanism in each event. Considering that aging and AD are related to cognitive impairment, here we discuss the involving these neurotrophic factors in the aging process and AD.

16.
An Acad Bras Cienc ; 87(2 Suppl): 1461-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26312426

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia and has become a severe public health issue. It is estimated that globally, 35.6% of people have some form of dementia. This number is expected to double by 2030, and possibly even triple by 2050. The disease is associated with deficits in cognition/memory and a reduced ability in coping with everyday life. Moreover, patients can experience behavioral alterations such as mood swings, depression and hallucinations. Therefore, it is common to find the presence of neuropsychiatric comorbidities such as depression, schizophrenia and bipolar disorder during the course or development of AD. These disorders can become severe enough to interfere with the patients daily functioning, and can worsen the course of the disease. However, little is known about the causal relationship between psychiatric comorbidities and AD, or the reasons for the predisposition of some individuals to such disorders. Therefore, the purpose of this review is to clarify the causal relationship between depression, schizophrenia and bipolar disorder with AD.


Subject(s)
Alzheimer Disease/psychology , Bipolar Disorder/psychology , Dementia/psychology , Schizophrenia , Comorbidity , Humans
17.
Brain Behav Immun ; 43: 54-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25019583

ABSTRACT

Oxidative stress and inflammation is likely to be a major step in the development of sepsis-associated encephalopathy (SAE) and long-term cognitive impairment. To date, it is not known whether brain inflammation and oxidative damage are a direct consequence of systemic inflammation or whether these events are driven by brain resident cells, such as microglia. Therefore, the aim of this study is to evaluate the effect of minocycline on behavioral and neuroinflammatory parameters in rats submitted to sepsis. Male Wistar rats were subjected to sepsis by cecal ligation and puncture (CLP). The animals were divided into sham-operated (Sham+control), sham-operated plus minocycline (sham+MIN), CLP (CLP+control) and CLP plus minocycline (CLP+MIN) (100 µg/kg, administered as a single intracerebroventricular (ICV) injection). Some animals were killed 24h after surgery to assess the breakdown of the blood brain barrier, cytokine levels, oxidative damage to lipids (TBARS) and proteins in the hippocampus. Some animals were allowed to recover for 10 days when step-down inhibitory avoidance and open-field tasks were performed. Treatment with minocycline prevented an increase in markers of oxidative damage and inflammation in the hippocampus after sepsis. This was associated with an improvement in long-term cognitive performance. In conclusion, we demonstrated that the inhibition of the microglia by an ICV injection of minocycline was able to decrease acute brain oxidative damage and inflammation as well as long-term cognitive impairment in sepsis survivors.


Subject(s)
Brain/metabolism , Cognition Disorders/etiology , Inflammation/metabolism , Microglia/metabolism , Sepsis/complications , Animals , Avoidance Learning/physiology , Cognition Disorders/metabolism , Cytokines/blood , Hippocampus/metabolism , Male , Oxidative Stress/physiology , Rats , Rats, Wistar , Sepsis/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
18.
Synapse ; 68(10): 474-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24978930

ABSTRACT

Sepsis is defined as the host's reaction to infection and it is characterized by a systemic inflammatory response with important clinical implications. Central nervous system dysfunction secondary to sepsis is associated with local generation of pro- and anti-inflammatory cytokines, impaired cerebral microcirculation, disturbance of neurotransmitters, apoptosis, and cognitive impairment. It is known that during the process of learning and memory formation several pathways are involved such as dopaminergic and cholinergic systems. Thus, the objective of this study is to evaluate the neuronal calcium sensor (NCS-1) and dopamine-cAMP regulated phosphoprotein of 32,000 kDa (DARPP-32) expression as well as brain-derived neurotrophic factor (BDNF) and nerve growth factor (NGF) levels in prefrontal cortex and hippocampus of rats 12, 24, and 48 h after sepsis induction. To this aim, we used sham-operated Wistar rats or submitted to the cecal ligation and perforation procedure. After 12 and 24 h, there was an increase of NGF levels in hippocampus; and up to 48 h, a decrease of NCS-1 expression in prefrontal cortex, a decrease of BDNF levels in hippocampus and an increase of NGF levels in hippocampus. In conclusion, we believe that the low expression of NCS-1 in prefrontal cortex and low levels of BDNF in hippocampus may be associated with the pathophysiology of cognitive impairment during sepsis and a putative role of the dopaminergic system.


Subject(s)
Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Hippocampus/metabolism , Nerve Growth Factors/metabolism , Neuronal Calcium-Sensor Proteins/metabolism , Neuropeptides/metabolism , Prefrontal Cortex/metabolism , Sepsis/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Male , Nerve Growth Factor/metabolism , Rats, Wistar , Time Factors
19.
Mol Neurobiol ; 49(2): 1069-76, 2014 04.
Article in English | MEDLINE | ID: mdl-24234155

ABSTRACT

Sepsis is defined as the host's reaction to infection and characterised by a systemic inflammatory response with important clinical implications. Central nervous system dysfunction secondary to sepsis is associated with local generation of pro- and anti-inflammatory cytokines, impaired cerebral microcirculation, an imbalance of neurotransmitters, apoptosis and cognitive impairment. It's known that the IL-1ß is one of the first cytokines to be altered. Thus, the objective of this study was to evaluate the role of IL-1ß in cognitive parameters in brain tissue through the use of an IL-1ß (IL-1ra) receptor antagonist up to 10 days and to assess blood-brain barrier permeability, cytokine levels, oxidative parameters and energetic metabolism up to 24 h, after sepsis induction. To this aim, we used sham-operated Wistar rats or submitted to the cecal ligation and perforation (CLP) procedure. Immediately after, the animals received one dose of 10 µg of IL-1ra. After 24 h, the rats were killed and were evaluated for biochemical parameters in the pre-frontal cortex, hippocampus and striatum. After 10 days, the animals were submitted to the habituation to the open field and step-down inhibitory avoidance task. We observed that the use of IL-1ra reverted the increase of blood-brain barrier permeability in the pre-frontal cortex, hippocampus and striatum; the increase of IL-1ß, IL1-6 and TNF-α levels in the pre-frontal cortex and striatum; the decrease of complex I activity in the pre-frontal, hippocampus and striatum; the increase of oxidative parameters in pre-frontal cortex, hippocampus and striatum; and cognitive impairment. In conclusion, the results observed in this study reinforce the role of acute brain inflammatory response, in particular, the IL1ß response, in the cognitive impairment associated with sepsis.


Subject(s)
Cognition Disorders/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , Sepsis/metabolism , Animals , Avoidance Learning/physiology , Cognition Disorders/pathology , Male , Oxidative Stress/physiology , Rats , Rats, Wistar , Sepsis/psychology
20.
Mol Neurobiol ; 48(3): 581-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23559405

ABSTRACT

Maple syrup urine disease (MSUD) is a neurometabolic disorder caused by deficiency of the activity of the mitochondrial enzyme complex branched-chain α-keto acid dehydrogenase leading to accumulation of the branched-chain amino acids (BCAA) and their corresponding branched-chain α-keto acids. In this study, we examined the effects of acute and chronic administration of BCAA on protein levels and mRNA expression of nerve growth factor (NGF) considering that patients with MSUD present neurological dysfunction and cognitive impairment. Considering previous observations, it is suggested that oxidative stress may be involved in the pathophysiology of the neurological dysfunction of MSUD. We also investigated the influence of antioxidant treatment (N-acetylcysteine and deferoxamine) in order to verify the influence of oxidative stress in the modulation of NGF levels. Our results demonstrated decreased protein levels of NGF in the hippocampus after acute and chronic administration of BCAA. In addition, we showed a significant decrease in the expression of ngf in the hippocampus only following acute administration in 10-day-old rats. Interestingly, antioxidant treatment was able to prevent the decrease in NGF levels by increasing ngf expression. In conclusion, the results suggest that BCAA is involved in the regulation of NGF in the developing rat. Thus, it is possible that alteration of neurotrophin levels during brain maturation could be of pivotal importance in the impairment of cognition provoked by BCAA. Moreover, the decrease in NGF levels was prevented by antioxidant treatment, reinforcing that the hypothesis of oxidative stress can be an important pathophysiological mechanism underlying the brain damage observed in MSUD.


Subject(s)
Amino Acids, Branched-Chain/administration & dosage , Amino Acids, Branched-Chain/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Nerve Growth Factor/metabolism , Animals , Antioxidants/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Gene Expression Regulation/drug effects , Male , Neostriatum/drug effects , Neostriatum/metabolism , Nerve Growth Factor/genetics , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL