Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(15)2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37569815

ABSTRACT

Alzheimer's disease (AD) is the leading cause of dementia in older adults, having a significant global burden and increasing prevalence. Current treatments for AD only provide symptomatic relief and do not cure the disease. Physical activity has been extensively studied as a potential preventive measure against cognitive decline and AD. Recent research has identified a hormone called irisin, which is produced during exercise, that has shown promising effects on cognitive function. Irisin acts on the brain by promoting neuroprotection by enhancing the growth and survival of neurons. It also plays a role in metabolism, energy regulation, and glucose homeostasis. Furthermore, irisin has been found to modulate autophagy, which is a cellular process involved in the clearance of protein aggregates, which are a hallmark of AD. Additionally, irisin has been shown to protect against cell death, apoptosis, oxidative stress, and neuroinflammation, all of which are implicated in AD pathogenesis. However, further research is needed to fully understand the mechanisms and therapeutic potential of irisin in AD. Despite the current gaps in knowledge, irisin holds promise as a potential therapeutic target for slowing cognitive decline and improving quality of life in AD patients.


Subject(s)
Alzheimer Disease , Healthy Aging , Aged , Humans , Alzheimer Disease/metabolism , Fibronectins/metabolism , Neuroprotection , Quality of Life
2.
Nutr Rev ; 81(10): 1310-1320, 2023 09 11.
Article in English | MEDLINE | ID: mdl-36779324

ABSTRACT

CONTEXT: A recent US national survey of the health status of the male transgender population has raised awareness about the little-studied relationship between testosterone hormone therapy in transgender men and cardiovascular outcomes. OBJECTIVE: The aim of this systematic review was to assess the relationship between cross-sex hormone therapy in transgender men and lipid profiles and cardiovascular risk. DATA SOURCES: The PubMed, SciELO, SpringerLink, and EBSCOhost databases were searched up to March 2021 for studies assessing the association between cross-sex hormone therapy and the incidence of outcomes related to cardiovascular disease in transgender men over 18 years of age . DATA EXTRACTION: Data extracted were sorted into clinical data (systolic, diastolic, and mean blood pressure), anthropometric data (body mass index, weight, waist circumference, fat mass, and lean mass), and biochemical data (triglycerides, total cholesterol, low-density lipoprotein cholesterol [LDL-C], high-density lipoprotein cholesterol [HDL-C], very low-density lipoprotein cholesterol [VLDL-C], and the HDL-C to LDL-C ratio). DATA ANALYSIS: Study quality was appraised independently by two reviewers using the Cochrane tools for assessment of methodological quality or risk of bias in nonrandomized studies, and the Newcastle-Ottawa Scale was applied. Of 735 studies identified, 11 were included in the review. Most studies reported no change in cholesterol or triglyceride levels after hormone treatment. A reduction in HDL-C levels was observed in 7 of 11 studies, although this alone cannot be considered a cardiovascular risk factor. Likewise, clinical and anthropometric findings showed no changes predictive of cardiovascular risk. CONCLUSIONS: Although these findings suggest that hormone therapy may lead to a decrease in HDL-C levels and an increase in LDL-C levels, they are insufficient to establish a relationship with cardiovascular disease. Furthermore, no significant effects on metabolic and anthropometric values were found. Further studies with higher quality and longer follow-up periods are needed to establish cardiovascular risk. SYSTEMATIC REVIEW REGISTRATION: PROSPERO registration number CRD 42020212560.


Subject(s)
Cardiovascular Diseases , Transgender Persons , Humans , Male , Adult , Adolescent , Cholesterol, LDL , Cardiovascular Diseases/epidemiology , Cholesterol , Triglycerides , Cholesterol, HDL , Gonadal Steroid Hormones , Risk Factors
3.
J Appl Microbiol ; 134(1)2023 Jan 23.
Article in English | MEDLINE | ID: mdl-36724248

ABSTRACT

AIMS: The protective effects of Bacillus amyloliquefaciens(CCT7935), Bacillus subtilis(CCT7935), Bacillus licheniformis (CCT 7836), and Bacillus coagulans (CCT 0199) against lipopolysaccharide (LPS)-induced intestinal inflammation were investigated. METHODS AND RESULTS: Male Swiss mice were assigned into six groups: control group, LPS group, LPS + B. subtilis (CCT7935) group, LPS +   B. licheniformis (CCT 7836) group, LPS +   B. amyloliquefaciens (CCT7935) group, and LPS   + B. coagulans (CCT 0199) group. Each mouse of the groups Bacillus received 1 × 109 colony-forming units of Bacillus once daily by oral gavage during 30 days. Twenty-four hours after the last dose of Bacillus, all groups, except the control group, were intraperitoneally injected with LPS in the single dose of 15 mg kg-1. The mice were euthanized 24 h after the LPS administration. Histological alterations, myeloperoxidase activity, and nitrite levels were analyzed in the gut of mice and the inflammatory cytokines were analyzed in the gut and in the blood. The results demonstrate that the mice challenged with LPS presented the villi shortened and damaged, which were significantly protected by B. coagulans and B. amyloliquefaciens. Furthermore, all Bacillus tested were effective in preventing against the increase of myeloperoxidase activity, while B. amyloliquefaciens and B. subtilis prevented the increase of nitrite and IL-1ß levels in the gut of mice induced with LPS was decreased only B. subtilis. LPS also elevated the IL-1 ß, IL-6, and IL-10 levels in the blood, and these alterations were significantly suppressed by Bacillus, especially by B. subtilis. CONCLUSIONS: The study suggests that the Bacillus investigated in this study might be effective therapeutic agents for preventing intestinal inflammation, because they decrease the inflammatory process an protect against tissue damage.


Subject(s)
Bacillus , Probiotics , Animals , Mice , Male , Lipopolysaccharides , Peroxidase , Nitrites , Probiotics/pharmacology , Inflammation/chemically induced , Inflammation/prevention & control
4.
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
5.
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
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.
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
8.
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
9.
Mol Neurobiol ; 57(11): 4451-4466, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32743736

ABSTRACT

Sepsis causes organ dysfunction due to an infection, and it may impact the central nervous system. Neuroinflammation and oxidative stress are related to brain dysfunction after sepsis. Both processes affect microglia activation, neurotrophin production, and long-term cognition. Fish oil (FO) is an anti-inflammatory compound, and lipoic acid (LA) is a universal antioxidant substance. They exert neuroprotective roles when administered alone. We aimed at determining the effect of FO+LA combination on microglia activation and brain dysfunction after sepsis. Microglia cells from neonatal pups were co-treated with lipopolysaccharide (LPS) and FO or LA, alone or combined, for 24 h. Cytokine levels were measured. Wistar rats were subjected to sepsis by cecal ligation and perforation (CLP) and treated orally with FO, LA, or FO+LA. At 24 h after surgery, the hippocampus, prefrontal cortex, and total cortex were obtained and assayed for levels of cytokines, myeloperoxidase (MPO) activity, protein carbonyls, superoxide dismutase (SOD), and catalase (CAT) activity. At 10 days after surgery, brain-derived neurotrophic factor (BDNF) levels were determined and behavioral tests were performed. The combination diminished in vitro levels of pro-inflammatory cytokines. The combination reduced TNF-α in the cortex, IL-1ß in the prefrontal cortex, as well as MPO activity, and decreased protein carbonyls formation in all structures. The combination enhanced catalase activity in the prefrontal cortex and hippocampus, elevated BDNF levels in all structures, and prevented behavioral impairment. In summary, the combination was effective in preventing cognitive damage by reducing neuroinflammation and oxidative stress and increasing BDNF levels.


Subject(s)
Brain/pathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Fish Oils/pharmacology , Inflammation/pathology , Oxidative Stress/drug effects , Sepsis/complications , Thioctic Acid/pharmacology , Animals , Brain/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Catalase/metabolism , Cells, Cultured , Cytokines/metabolism , Female , Inflammation/complications , Kaplan-Meier Estimate , Memory Disorders/complications , Microglia/drug effects , Microglia/metabolism , Open Field Test , Peroxidase/metabolism , Protein Carbonylation/drug effects , Rats, Wistar , Superoxide Dismutase/metabolism
10.
Nutrition ; 70: 110417, 2020 02.
Article in English | MEDLINE | ID: mdl-30867119

ABSTRACT

OBJECTIVES: Sepsis is a severe organic dysfunction caused by an infection that affects the normal regulation of several organ systems, including the central nervous system. Inflammation and oxidative stress play crucial roles in the development of brain dysfunction in sepsis. The aim of this study was to determine the effect of a fish oil (FO)-55-enriched lipid emulsion as an important anti-inflammatory compound on brain dysfunction in septic rats. METHODS: Wistar rats were subjected to sepsis by cecal ligation and perforation (CLP) or sham (control) and treated orally with FO (600 µL/kg after CLP) or vehicle (saline; sal). Animals were divided into sham+sal, sham+FO, CLP+sal and CLP+FO groups. At 24 h and 10 d after surgery, the hippocampus, prefrontal cortex, and total cortex were obtained and assayed for levels of interleukin (IL)-1ß and IL-10, blood-brain barrier permeability, nitrite/nitrate concentration, myeloperoxidase activity, thiobarbituric acid reactive species formation, protein carbonyls, superoxide dismutase and catalase activity, and brain-derived neurotrophic factor levels. Behavioral tasks were performed 10 d after surgery. RESULTS: FO reduced BBB permeability in the prefrontal cortex and total cortex of septic rats, decreased IL-1ß levels and protein carbonylation in all brain structures, and diminished myeloperoxidase activity in the hippocampus and prefrontal cortex. FO enhanced brain-derived neurotrophic factor levels in the hippocampus and prefrontal cortex and prevented cognitive impairment. CONCLUSIONS: FO diminishes the negative effect of polymicrobial sepsis in the rat brain by reducing inflammatory and oxidative stress markers.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cognitive Dysfunction/prevention & control , Fish Oils/pharmacokinetics , Oxidative Stress/drug effects , Sepsis/psychology , Animals , Biomarkers/metabolism , Blood-Brain Barrier/drug effects , Brain/drug effects , Cecal Diseases/complications , Cecal Diseases/microbiology , Cecum/blood supply , Cecum/microbiology , Cognitive Dysfunction/microbiology , Disease Models, Animal , Emulsions , Frontal Lobe/drug effects , Interleukin-1beta/metabolism , Intestinal Perforation/complications , Intestinal Perforation/microbiology , Ligation/adverse effects , Male , Permeability , Protein Carbonylation/drug effects , Rats , Rats, Wistar , Sepsis/etiology , Sepsis/microbiology
11.
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
12.
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
13.
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
14.
Article in English | MEDLINE | ID: mdl-28336494

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder and the most common type of age-related dementia. Cognitive decline, beta-amyloid (Aß) accumulation, neurofibrillary tangles, and neuroinflammation are the main pathophysiological characteristics of AD. Minocycline is a tetracycline derivative with anti-inflammatory properties that has a neuroprotective effect. The aim of this study was to evaluate the effect of minocycline on memory, neurotrophins and neuroinflammation in an animal model of AD induced by the administration of Aß (1-42) oligomer. Male BALB/c mice were treated with minocycline (50mg/kg) via the oral route for a total of 17days, 24h after intracerebroventricular administration of Aß (1-42) oligomer. At the end of this period, was performed the radial maze test, and 24h after the last minocycline administration, serum was collected and the cortex and hippocampus were dissected for biochemical analysis. The administration of minocycline reversed the memory impairment caused by Aß (1-42). In the hippocampus, minocycline reversed the increases in the levels of interleukin (IL-1ß), Tumor Necrosis Factor- alpha (TNF-α) and, IL-10 caused by Aß (1-42). In the cortex, AD-like model increase the levels of IL-1ß, TNF-α and, IL-4. Minocycline treatment reversed this. In the serum, Aß (1-42) increased the levels of IL-1ß and IL-4, and minocycline was able to reverse this action, but not to reverse the decrease of IL-10 levels. Minocycline also reversed the increase in the levels of Brain-derived neurotrophic factor (BDNF) in the hippocampus caused by Aß (1-42), and reduced Nerve Growth Factor (NGF) increases in the total cortex. Therefore, our results indicate that minocycline causes improvements in the spatial memory, and cytokine levels were correlated with this effect in the brain it. Besides this, minocycline reduced BDNF and NGF levels, highlighting the promising effects of minocycline in treating AD-like dementia.


Subject(s)
Amyloid beta-Peptides/toxicity , Brain/drug effects , Inflammation/prevention & control , Memory Disorders/prevention & control , Minocycline/pharmacology , Neuroprotective Agents/pharmacology , Peptide Fragments/toxicity , Amyloid beta-Peptides/administration & dosage , Animals , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Inflammation/blood , Inflammation/chemically induced , Inflammation/metabolism , Infusions, Intraventricular , Interleukin-10/blood , Interleukin-10/metabolism , Interleukin-1beta/blood , Interleukin-1beta/metabolism , Interleukin-4/blood , Interleukin-4/metabolism , Male , Maze Learning/drug effects , Memory Disorders/blood , Memory Disorders/chemically induced , Memory Disorders/metabolism , Mice , Peptide Fragments/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
15.
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
16.
Behav Brain Res ; 302: 35-43, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26748256

ABSTRACT

d-Galactose (d-gal) is a reducing sugar that can be used to mimic the characteristics of aging in rodents; however, the effects of d-gal administration by oral route are not clear. Therefore, the aim of this study was to elucidate if the oral administration of d-gal induces cognitive impairments, neuronal loss, and oxidative damage, mimicking an animal model of aging. Male adult Wistar rats (4 months old) received d-gal (100mg/kg) via the oral route for a period of 1, 2, 4, 6 or 8 weeks. The results showed cognitive impairments in the open-field test in the 4th and 6th weeks after d-gal administration, as well as an impairment in spatial memory in the radial maze test after the 6th week of d-gal administration. The results indicated increase of levels of thiobarbituric acid reactive species-TBARS-and carbonyl group content in the prefrontal cortex from the 4th week, and in all weeks of d-gal administration, respectively. An increase in the levels of TBARS and carbonyl group content was observed in the hippocampus over the entire period of d-gal treatment. In the 8th week of d-gal administration, we also observed reductions in synaptophysin and TAU protein levels in the prefrontal cortex. Thus, d-gal given by oral route caused cognitive impairments which were accompanied by oxidative damage. Therefore, these results indicate that orally administered d-gal can induce the behavioral and neurochemical alterations that are observed in the natural aging process. However, oral d-gal effect in rats deserve further studies to be better described.


Subject(s)
Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Galactose/administration & dosage , Oxidative Stress/drug effects , Administration, Oral , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cognition Disorders/pathology , Disease Models, Animal , Exploratory Behavior/drug effects , Follow-Up Studies , Galactose/pharmacology , Male , Maze Learning/drug effects , Protein Carbonylation/drug effects , Rats , Rats, Wistar , Reaction Time/drug effects , Synaptophysin/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Time Factors , tau Proteins/metabolism
17.
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.

SELECTION OF CITATIONS
SEARCH DETAIL
...