Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters

Database
Country/Region as subject
Language
Affiliation country
Publication year range
1.
Brain Res ; 1784: 147845, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35219720

ABSTRACT

Essential oils (EO) are plant extracts widely used for various pharmacological applications and their antioxidant and anti-inflammatory effects have received a lot of attention because they hold the potential to reduce oxidative stress, and neuroinflammation, alterations involved in the pathophysiology of major depressive disorder. This study examined the benefits of administration of flower EO of the Tagetes minuta (10 and 50 mg/kg, intragastric route) in attenuating behavioral, neurochemical, and neuroendocrine changes in animal models of depressive-like behavior induced by acute restraint stress and lipopolysaccharide (0.83 mg/kg, intraperitoneally). We demonstrated that the treatment of mice with flower EO of the T. minuta reversed the depressive-like behavior induced by stress or inflammatory challenge in mice. This effect is most likely due to the reversal of oxidative stress in the hippocampus of mice, the decrease in plasma corticosterone levels, and restoration of the mRNA levels of brain-derived neurotrophic factor, phosphatidylinositol-3-kinase, protein kinase B, and extracellular signal-regulated kinase 2. As an outcome, flower EO of the T. minuta has promising antidepressant properties and could be considered for new therapeutic strategies for major depressive disorder.


Subject(s)
Depressive Disorder, Major , Oils, Volatile , Tagetes , Animals , Behavior, Animal , Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Depression/metabolism , Depressive Disorder, Major/metabolism , Disease Models, Animal , Flowers/metabolism , Hippocampus/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Oils, Volatile/metabolism , Oils, Volatile/pharmacology , Oils, Volatile/therapeutic use , Oxidative Stress , Proto-Oncogene Proteins c-akt/metabolism , Tagetes/metabolism
2.
Neurochem Res ; 46(3): 535-549, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33548035

ABSTRACT

Extensive data have reported the involvement of oxidative stress in the pathogenesis of neuropsychiatric disorders, prompting the pursuit of antioxidant molecules that could become adjuvant pharmacological agents for the management of oxidative stress-associated disorders. The 3-[(4-chlorophenyl)selanyl]-1-methyl-1H-indole (CMI) has been reported as an antioxidant and immunomodulatory compound that improves depression-like behavior and cognitive impairment in mice. However, the exact effect of CMI on specific brain cells is yet to be studied. In this context, the present study aimed to evaluate the antioxidant activity of CMI in H2O2-induced oxidative stress on human dopaminergic neuroblastoma cells (SH-SY5Y) and to shed some light into its possible mechanism of action. Our results demonstrated that the treatment of SH-SY5Y cells with 4 µM CMI protected them against H2O2 (343 µM)-induced oxidative stress. Specifically, CMI prevented the increased number of reactive oxygen species (ROS)-positive cells induced by H2O2 exposure. Furthermore, CMI treatment increased the levels of reduced glutathione in SH-SY5Y cells. Molecular docking studies demonstrated that CMI might interact with enzymes involved in glutathione metabolism (i.e., glutathione peroxidase and glutathione reductase) and H2O2 scavenging (i.e., catalase). In silico pharmacokinetics analysis predicted that CMI might be well absorbed, metabolized, and excreted, and able to cross the blood-brain barrier. Also, CMI was not considered toxic overall. Taken together, our results suggest that CMI protects dopaminergic neurons from H2O2-induced stress by lowering ROS levels and boosting the glutathione system. These results will facilitate the clinical application of CMI to treat nervous system diseases associated with oxidative stress.


Subject(s)
Hydrogen Peroxide/toxicity , Indoles/pharmacology , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Selenium Compounds/pharmacology , Catalytic Domain , Cell Line, Tumor , Glutathione/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Humans , Indoles/chemistry , Indoles/metabolism , Indoles/pharmacokinetics , Molecular Docking Simulation , Neuroprotective Agents/chemistry , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Protein Binding , Reactive Oxygen Species/metabolism , Selenium Compounds/chemistry , Selenium Compounds/metabolism , Selenium Compounds/pharmacokinetics
3.
Brain Res ; 1741: 146880, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32417177

ABSTRACT

Bearing in mind that pain and major depressive disorder (MDD) often share biological pathways, this condition is classified as depression-pain syndrome. Mounting evidence suggests that oxidative stress is implicated in the pathophysiology of this syndrome. The development of effective pharmacological interventions for the depression-pain syndrome is of particular importance as clinical treatments for this comorbidity have shown limited efficacy. Therefore, the present study aimed to evaluate whether the 3,5-dimethyl-1-phenyl-4-(phenylselanyl)-1H-pyrazole (SePy) was able to reverse the depression-pain syndrome induced by intracerebroventricular (i.c.v) streptozotocin (STZ) in mice and the possible modulation of oxidative and nitrergic pathways in its effect. The treatment with SePy (1 and 10 mg/kg) administered intragastrically (i.g.) reversed the increased immobility time in the tail suspension test, decreased grooming time in the splash test, latency time to nociceptive response in the hot plate test, and the response frequency of Von Frey hair (VFH) stimulation induced by STZ (0.2 mg/4 µl/per mouse). Additionally, SePy (10 mg/kg, i.g.) reversed STZ-induced alterations in the levels of reactive oxygen species, nitric oxide, and lipid peroxidation and the superoxide dismutase and catalase activities in the prefrontal cortices (PFC) and hippocampi (HC) of mice. Treatment with SePy (10 mg/kg, i.g.) also reversed the STZ-induced increased expression of inducible nitric oxide synthase (iNOS) and glycogen synthase kinase 3 beta (GSK3ß) in the PFC and HC. An additional molecular docking investigation found that SePy binds to the active site of iNOS and GSK3ß. Altogether, these results indicate that the antidepressant-like effect of SePy is accompanied by decreased hyperalgesia and mechanical allodynia, which were associated with its antioxidant effect.


Subject(s)
Depression/drug therapy , Nitrosative Stress/drug effects , Oxidative Stress/drug effects , Pain/drug therapy , Pyrazoles/administration & dosage , Selenium/administration & dosage , Animals , Depression/chemically induced , Depression/metabolism , Glycogen Synthase Kinase 3 beta/chemistry , Glycogen Synthase Kinase 3 beta/metabolism , Injections, Intraventricular , Male , Mice , Nitric Oxide Synthase Type II/chemistry , Nitric Oxide Synthase Type II/metabolism , Nitrosative Stress/physiology , Oxidative Stress/physiology , Pain/chemically induced , Pain/metabolism , Pain Measurement/drug effects , Pain Measurement/methods , Protein Structure, Secondary , Streptozocin/administration & dosage , Streptozocin/toxicity
4.
Brain Behav Immun ; 84: 229-241, 2020 02.
Article in English | MEDLINE | ID: mdl-31837417

ABSTRACT

Psychiatric alterations are often found in patients with breast cancer even before the initiation of adjuvant therapy, resulting in a poor quality of life. It has become accepted that neuroinflammation and oxidative stress are involved in the pathophysiology of depression and cognitive impairment. Herein, we tested the hypothesis that treatment with the antioxidant and immunomodulatory selenium-containing compound 3-[(4-chlorophenyl)selanyl]-1-methyl-1H-indole (CMI)could attenuate behavioral and neurochemical alterations in a mammary (4T1) tumor model. Female BALB/c mice were subcutaneously inoculated with 4T1 cancer cells (1 × 105 cells/mice) or PBS. From days 14 to 20, mice received daily gavage with canola oil or CMI. On day 21, mice were submitted to behavioral tests followed by euthanasia. We found that CMI did not alter tumor growth, body weight, and body temperature in tumor-bearing mice. Importantly, treatment with CMI abrogated tumor-induced depression-like behavior and cognitive impairment. By the time CMI improved the behavioral alterations, it had reduced tumor-induced neuroinflammation (altered expression of NFκB, IL-1ß, TNF-α, IL-10, IDO, and COX-2) and oxidative stress (altered expression of iNOS and Nrf2, and levels of reactive species, nitric oxide, lipid peroxidation, and superoxide dismutase activity) in the prefrontal cortices and hippocampi of mice. A molecular docking approach suggested the ability of CMI to inhibit the activity of iNOS and COX-2. Together, our results indicate that CMI treatment may attenuate depression and cognitive impairment in 4T1 tumor-bearing mice, and be a groundbreaking strategy for the treatment of cancer-related psychiatric symptoms to improve the quality of life of cancer patients.


Subject(s)
Antioxidants , Breast Neoplasms , Cognitive Dysfunction , Depression , Indoles , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Breast Neoplasms/complications , Breast Neoplasms/drug therapy , Cognitive Dysfunction/drug therapy , Depression/drug therapy , Depression/etiology , Disease Models, Animal , Female , Humans , Indoles/pharmacology , Indoles/therapeutic use , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Oxidative Stress/drug effects , Quality of Life , Selenium Compounds
5.
J Psychiatr Res ; 120: 91-102, 2020 01.
Article in English | MEDLINE | ID: mdl-31654972

ABSTRACT

Oxidative stress and neuroinflammation are found both in diabetes mellitus and major depressive disorder (MDD). In addition to damage in peripheral organs, such as liver and kidney, diabetic patients have a higher risk of developing depression. In this sense, the objective of the present study was to characterize the antidepressant-like effect of a selenium-containing compound, the 1-methyl-3-(phenylselanyl)-1H-indole (MFSeI), in streptozotocin (STZ)-induced diabetic mice. STZ (200 mg/kg, i.p.) was used to induce diabetes mellitus type I, and after seven days, the administration of MFSeI (10 mg/kg, i.g.) was initiated and followed for the next 14 days. Twenty-four hours after the last administration of MFSeI, the behavioral tests were performed, followed by euthanasia. The treatment with MFSeI was able to reverse the hyperglycemia induced by STZ. MFSeI also decreased the plasma levels of biomarkers of liver and kidney damage. Importantly, MFSeI reversed the depression-like behavior induced by STZ in the tail suspension test and forced swimming test without promoting locomotor alterations. Furthermore, MFSeI reversed the increased levels of reactive species and lipid peroxidation in the prefrontal cortex (PFC), hippocampus (HC), liver, and kidney of STZ-treated mice. Treatment with MFSeI also decreased the expression of tumor necrosis factor-alpha, inducible nitric oxide synthase and indoleamine 2,3-dioxygenase, while increasing the expression of interleukin-10, insulin receptor substrate-1 and glucose transport-4 in the PFC and HC of mice. Taken together, the results indicate the effectiveness of MFSeI against depression-like behavior and central and peripheral complications caused by diabetes in mice.


Subject(s)
Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Depression/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Hyperglycemia/drug therapy , Indoles/pharmacology , Inflammation/drug therapy , Organoselenium Compounds/pharmacology , Animals , Depression/blood , Depression/immunology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/immunology , Hippocampus/drug effects , Hyperglycemia/blood , Hyperglycemia/immunology , Indoles/administration & dosage , Inflammation/blood , Inflammation/immunology , Kidney/drug effects , Liver/drug effects , Mice , Organoselenium Compounds/administration & dosage , Selenium
6.
Behav Brain Res ; 366: 96-107, 2019 07 02.
Article in English | MEDLINE | ID: mdl-30877027

ABSTRACT

In the last decades, selenium-containing compounds have received increasing attention due to their various biological and pharmacological properties. In the present study, we investigated the effects of 3-[(4-methoxyphenyl) selanyl]-2-phenylimidazo[1,2-a] pyridine (MPI; 1, 10 or 50 mg/kg, i.g.) on the acute restraint stress (ARS)-induced depressive- and anxiety-like behaviors in mice and its underlying mechanism of action. We used the open filed test, forced swimming test, and splash test to evaluate depressive-like behavior, and marble burying and elevated plus maze test to measure anxiety-like behavior. We found that MPI attenuated ARS-induced depressive- and anxiety-like behaviors in all behavioral tests, without having an effect in non-stressed mice. MPI prevented the increased in pro-inflammatory cytokines, indoleamine-2,3-dioxygenase (IDO) and inducible nitric oxide synthase (iNOS) expression in brain structures via canonical nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) down-regulation. Additionally, MPI prevented ARS-induced downregulation of brain-derived neurotrophic factor (BDNF), increased reactive oxygen/nitrogen species generation and lipid peroxidation in prefrontal cortex and hippocampus of mice. In addition, MPI blocked the downregulation of glucocorticoid receptors in the prefrontal cortex and hippocampus and reduced the increased circulating level of corticosterone in stressed mice. These results suggested that MPI showed antidepressant- and anxiolytic-like properties and the effects might be associated with the biological changes in the prefrontal cortex and hippocampus.


Subject(s)
Behavior, Animal/drug effects , Imidazoles/pharmacology , Selenium/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Anxiety/drug therapy , Anxiety/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Corticosterone/metabolism , Depression/drug therapy , Depression/metabolism , Disease Models, Animal , Hippocampus/drug effects , Imidazoles/metabolism , Lipid Peroxidation/drug effects , Male , Mice , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Prefrontal Cortex/drug effects , Pyridines/pharmacology , Receptors, Glucocorticoid/metabolism , Restraint, Physical , Selenium/metabolism , Stress, Psychological/metabolism
7.
Toxicol In Vitro ; 45(Pt 1): 194-201, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28923420

ABSTRACT

The improvement of in vitro embryo production by culture media supplementation has been a potential tool to increase blastocyst quality and development. Recently, lipid-core nanocapsules (LNC), which were developed for biomedical applications as a drug-delivery system, have demonstrated beneficial effects on in vitro embryo production studies. LNCs have a core composed of sorbitan monostearate dispersed in capric/caprylic triglyceride. Based on that, we firstly investigated if LNCs supplemented during in vitro oocyte maturation had affinity to the mineral oil placed over the top of the IVM media. Also, the effects of LNC supplementation in different concentrations (0; 0.94; 4.71; 23.56; 117.80 and 589.00µg/mL) during the in vitro maturation protocol were evaluated in oocytes and blastocysts by in vitro tests. LNCs seemed not to migrate to the mineral oil overlay during the in vitro oocyte maturation. Interestingly, LNCs did not show toxic effects in the oocyte in vitro maturation rate, cumulus cells expansion and oocyte viability. The highest LNCs concentration tested (589µg/mL) generated the lowest ROS and GSH levels, and reduced apoptosis rate when compared to the control. Additionally, toxic effects in embryo development and quality were not observed. The LNC supramolecular structure demonstrated to be a promising nanocarrier to deliver molecules in oocytes and embryos, aiming the improvement of the embryo in vitro development.


Subject(s)
Cattle/embryology , Embryo, Mammalian/drug effects , Embryonic Development/drug effects , Lipids/chemistry , Nanocapsules/toxicity , Animals , Embryo Culture Techniques/veterinary , Fertilization in Vitro/veterinary , In Vitro Oocyte Maturation Techniques/veterinary , Nanocapsules/chemistry
8.
J Psychopharmacol ; 31(9): 1263-1273, 2017 09.
Article in English | MEDLINE | ID: mdl-28661258

ABSTRACT

Organoselenium compounds and indoles have gained attention due to their wide range of pharmacological properties. Depression is a recurrent and disabling psychiatric illness and current evidences support that oxidative stress and neuroinflammation are mechanisms underlying the pathophysiology of this psychiatric condition. Here, we evaluated the effect of 3-((4-chlorophenyl)selanyl)-1-methyl-1H-indole (CMI) in lipopolysaccharide (LPS)-induced depressive-like behaviour, neuroinflammation and oxidative stress in male mice. CMI pre-treatment (20 and 50 mg/kg, intragastrically) significantly attenuated LPS (0.83 mg/kg, intraperitoneally)-induced depressive-like behaviour in mice by reducing the immobility time in the tail suspension test (TST) and forced swimming test (FST). CMI pre-treatment ameliorated LPS-induced neuroinflammation by reducing the levels of interleukin (IL)-1ß, IL-4 and IL-6 in the hippocampus and prefrontal cortex, as well as markers of oxidative damage. Additionally, we investigated the toxicological effects of CMI (200 mg/kg, i.g.) in the liver, kidney and brain through determination of the activity of aspartate aminotransferase (AST), alanine aminotransferase (ALT), δ-aminolevulinate dehydratase (δ-ALA-D) and creatinine levels. These biomarkers were not modified, indicating the possible absence of neuro-, hepato- and nephrotoxic effects. Our results suggest that CMI could be a therapeutic approach for the treatment of depression and other neuropsychiatric disorders associated with inflammation and oxidative stress.


Subject(s)
Antidepressive Agents/pharmacology , Inflammation/drug therapy , Lipopolysaccharides/pharmacology , Oxidative Stress/drug effects , Selenium/pharmacology , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Depression/drug therapy , Depression/metabolism , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukin-4/metabolism , Interleukin-6/metabolism , Male , Mice , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Swimming , Tumor Necrosis Factor-alpha/metabolism
9.
J Biosci ; 37(2): 227-32, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22581328

ABSTRACT

Neuropeptide Y (NPY) is considered the major stimulant for food intake in mammals and fish. Previous results indicate that NPY is involved in the feeding behaviour of the Brazilian flounder, Paralichthys orbignyanus. In this study, we evaluated hypothalamic NPY expression before (-2 h), during (0 h) and after feeding (+2 h) in two independent experiments: (1) during a normal feeding schedule and (2) in fish fasted for 2 weeks. During normal feeding, changes in the levels of NPY mRNA were periprandial, with expression levels being significantly elevated at meal time (P less than 0.05) and significantly reduced 2 h later (P less than 0.05). Comparing the fasting and unfasted groups, NPY mRNA levels were significantly higher (P less than 0.05) at -2 h and +2 h in the fasting group, but there was no difference at 0 h. In addition, the higher NPY mRNA levels that were observed in the fasting group were maintained throughout the sampling period. In summary, our results show that NPY expression was associated with meal time (0 h) in food intake regulation.


Subject(s)
Eating/physiology , Flounder/physiology , Neuropeptide Y/biosynthesis , Animals , Fasting/physiology , Flounder/metabolism , Gene Expression , Hypothalamus/metabolism , Neuropeptide Y/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL