Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 67
1.
J Toxicol Environ Health A ; 87(4): 166-184, 2024 Feb 16.
Article En | MEDLINE | ID: mdl-38073470

Copper (Cu) is a naturally occurring metal with essential micronutrient properties. However, this metal might also pose increased adverse environmental and health risks due to industrial and agricultural activities. In Brazil, the maximum allowable concentration of Cu in drinking water is 2 mg/L. Despite this standard, the impact of such concentrations on aquatic organisms remains unexplored. This study aimed to evaluate the toxicity of CuSO4 using larval zebrafish at environmentally relevant concentrations. Zebrafish (Danio rerio) larvae at 72 hr post-fertilization (hpf) were exposed to nominal CuSO4 concentrations ranging from 0.16 to 48 mg/L to determine the median lethal concentration (LC50), established at 8.4 mg/L. Subsequently, non-lethal concentrations of 0.16, 0.32, or 1.6 mg/L were selected for assessing CuSO4 -induced toxicity. Morphological parameters, including body length, yolk sac area, and swim bladder area, were adversely affected by CuSO4 exposure, particularly at 1.6 mg/L (3.31 mm ±0.1, 0.192 mm2 ±0.01, and 0.01 mm2 ±0.05, respectively). In contrast, the control group exhibited values of 3.62 mm ±0.09, 0.136 mm2 ±0.013, and 0.3 mm2 ±0.06, respectively. Behavioral assays demonstrated impairments in escape response and swimming capacity, accompanied by increased levels of reactive oxygen species (ROS) and lipid peroxidation. In addition, decreased levels of non-protein thiols and reduced cellular viability were noted. Data demonstrated that exposure to CuSO4 at similar concentrations as those permitted in Brazil for Cu adversely altered morphological, biochemical, and behavioral endpoints in zebrafish larvae. This study suggests that the permissible Cu concentrations in Brazil need to be reevaluated, given the potential enhanced adverse health risks of exposure to environmental metal contamination.


Copper , Water Pollutants, Chemical , Animals , Copper/toxicity , Zebrafish/physiology , Larva , Brazil , Lethal Dose 50 , Water Pollutants, Chemical/toxicity , Embryo, Nonmammalian
2.
Cell Chem Biol ; 31(2): 249-264.e7, 2024 Feb 15.
Article En | MEDLINE | ID: mdl-37944523

Iron overload, characterized by accumulation of iron in tissues, induces a multiorgan toxicity whose mechanisms are not fully understood. Using cultured cell lines, Caenorhabditis elegans, and mice, we found that ferroptosis occurs in the context of iron-overload-mediated damage. Exogenous oleic acid protected against iron-overload-toxicity in cell culture and Caenorhabditis elegans by suppressing ferroptosis. In mice, oleic acid protected against FAC-induced liver lipid peroxidation and damage. Oleic acid changed the cellular lipid composition, characterized by decreased levels of polyunsaturated fatty acyl phospholipids and decreased levels of ether-linked phospholipids. The protective effect of oleic acid in cells was attenuated by GW6471 (PPAR-α antagonist), as well as in Caenorhabditis elegans lacking the nuclear hormone receptor NHR-49 (a PPAR-α functional homologue). These results highlight ferroptosis as a driver of iron-overload-mediated damage, which is inhibited by oleic acid. This monounsaturated fatty acid represents a potential therapeutic approach to mitigating organ damage in iron overload individuals.


Ferroptosis , Iron Overload , Animals , Mice , Caenorhabditis elegans , Oleic Acid/pharmacology , Peroxisome Proliferator-Activated Receptors , Iron Overload/drug therapy , Iron , Phospholipid Ethers
3.
Anal Biochem ; 687: 115445, 2024 04.
Article En | MEDLINE | ID: mdl-38135241

REAP+ is an enhanced version of the rapid, efficient, and practical (REAP) method designed for the isolation of nuclear fractions. This improved version, REAP+, enables fast and effective extraction of mitochondria, cytoplasm, and nuclei. The mechanical cell disruption process has been optimized to cerebral tissues, snap-frozen liver, and HT22 cells with remarkable fraction enrichment. REAP+ is well-suited for samples containing minimal protein quantities, such as mouse hippocampal slices. The method was validated by Western blot and marker enzyme activities, such as LDH and G6PDH for the cytoplasmic fraction and succinate dehydrogenase and cytochrome c oxidase for the mitochondrial fraction. One of the outstanding features of this method is its rapid execution, yielding fractions within 15 min, allowing for simultaneous preparation of multiple samples. In essence, REAP+ emerges as a swift, efficient, and practical technique for the concurrent isolation of nuclei, cytoplasm, and mitochondria from various cell types and tissues. The method would be suitable to study the multicompartment translocation of proteins, such as metabolic enzymes and transcription factors migrating from cytosol to the mitochondria and nuclei. Moreover, its compatibility with small samples, such as hippocampal slices, and its potential applicability to human biopsies, highlights the potential application in medical research.


Cell Nucleus , Mitochondria , Humans , Mice , Animals , Cell Fractionation/methods , Mitochondria/metabolism , Cytoplasm/metabolism , Cell Nucleus/metabolism , Cytosol/metabolism , Subcellular Fractions/metabolism
4.
Int J Mol Sci ; 24(16)2023 Aug 09.
Article En | MEDLINE | ID: mdl-37628801

Huntington's disease (HD) is a neurodegenerative genetic disorder characterized by motor, psychiatric, cognitive, and peripheral symptoms without effective therapy. Evidence suggests that lifestyle factors can modulate disease onset and progression, and environmental enrichment (EE) has emerged as a potential approach to mitigate the progression and severity of neurodegenerative processes. Wild-type (WT) and yeast artificial chromosome (YAC) 128 mice were exposed to different EE conditions. Animals from cohort 1 were exposed to EE between postnatal days 21 and 60, and animals from cohort 2 were exposed to EE between postnatal days 60 and 120. Motor and non-motor behavioral tests were employed to evaluate the effects of EE on HD progression. Monoamine levels, hippocampal cell proliferation, neuronal differentiation, and dendritic arborization were also assessed. Here we show that EE had an antidepressant-like effect and slowed the progression of motor deficits in HD mice. It also reduced monoamine levels, which correlated with better motor performance, particularly in the striatum. EE also modulated neuronal differentiation in the YAC128 hippocampus. These results confirm that EE can impact behavior, hippocampal neuroplasticity, and monoamine levels in YAC128 mice, suggesting this could be a therapeutic strategy to modulate neuroplasticity deficits in HD. However, further research is needed to fully understand EE's mechanisms and long-term effects as an adjuvant therapy for this debilitating condition.


Heredodegenerative Disorders, Nervous System , Huntington Disease , Animals , Mice , Huntington Disease/genetics , Huntington Disease/therapy , Amines , Cell Proliferation , Combined Modality Therapy
5.
Inflammation ; 45(5): 1895-1910, 2022 Oct.
Article En | MEDLINE | ID: mdl-35727396

The association between chronic kidney disease (CKD) and pulmonary pathophysiological changes is well stablished. Nevertheless, the effects of aerobic exercise (AE) on lungs of CKD need further clarification. Thus, Swiss mice were divided in control, AE, CKD, and CKD + AE groups. CKD was induced by 0.2% adenine intake during 8 weeks (4 weeks of CKD induction and 4 weeks of AE). AE consisted in running on treadmill, at moderate intensity, 30 min/day, 5 days/week, during 4 weeks. Twenty-four hours after the last training day, functional capacity test was performed, and 48 h after the test, mice were euthanized. CKD mice showed a significant increase in urine output, serum urea, and creatinine concentrations, and decreased body weight and urine density, besides oxidative damage (p = 0.044), edema area (p < 0.001), leukocyte infiltration (p = 0.040), and collagen area in lung tissue (p = 0.004). AE resulted in an increase of distance traveled (p = 0.049) and maximum speed (p = 0.046), increased activity of catalase (p = 0.031) and glutathione peroxidase (p = 0.048) in lungs, increased levels of nitric oxide (NOx) in serum (p = 0.001) and bronchoalveolar lavage fluid (p = 0.047), and decreased kidney histological injury (p = 0.018) of CKD mice. However, AE also increased oxidative damage (p = 0.003) and did not change collagen content or perivascular edema in lungs (p > 0.05) of CKD mice. Therefore, AE attenuated kidney injury and improved antioxidants defenses in lungs. Despite no significant changes in pulmonary damage, AE significantly improved physical performance in CKD mice.


Antioxidants , Renal Insufficiency, Chronic , Adenine/pharmacology , Animals , Antioxidants/pharmacology , Catalase/metabolism , Creatinine , Glutathione Peroxidase , Kidney/pathology , Lung/metabolism , Mice , Nitric Oxide , Oxidative Stress , Physical Functional Performance , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/pathology , Urea/pharmacology
6.
Metab Brain Dis ; 37(5): 1597-1608, 2022 06.
Article En | MEDLINE | ID: mdl-35435610

Cholecalciferol deficiency has been associated with stress-related psychiatric disorders, particularly depression. Therefore, the present study investigated the antidepressant-like effect of cholecalciferol in female mice and the possible role of the serotonergic system in this response. The ability of cholecalciferol to elicit an antidepressant-like effect and to modulate serotonin levels in the hippocampus and prefrontal cortex of mice subjected to chronic unpredictable stress (CUS) was also investigated. The administration of cholecalciferol (2.5, 7.5, and 25 µg/kg, p.o.) for 7 days, similar to fluoxetine (10 mg/kg, p.o., serotonin reuptake inhibitor), reduced the immobility time in the tail suspension test, without altering the locomotor performance in the open-field test. Moreover, the administration of p-chlorophenylalanine methyl ester (PCPA - 100 mg/kg, i.p., for 4 days, a selective inhibitor of tryptophan hydroxylase, involved in the serotonin synthesis) abolished the antidepressant-like effect of cholecalciferol and fluoxetine in the tail suspension test, demonstrating the involvement of serotonergic system. Additionally, CUS protocol (21 days) induced depressive-like behavior in the tail suspension test and decreased serotonin levels in the prefrontal cortex and hippocampus of mice. Conversely, the administration of cholecalciferol and fluoxetine in the last 7 days of CUS protocol completely abolished the stress-induced depressive-like phenotype. Cholecalciferol was also effective to abrogate CUS-induced reduction on serotonin levels in the prefrontal cortex, but not in the hippocampus. Our results indicate that cholecalciferol has an antidepressant-like effect in mice by modulating the serotonergic system and support the assumption that cholecalciferol may have beneficial effects for the management of depression.


Fluoxetine , Serotonin , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Behavior, Animal , Cholecalciferol/pharmacology , Cholecalciferol/therapeutic use , Depression/drug therapy , Female , Fluoxetine/pharmacology , Fluoxetine/therapeutic use , Hindlimb Suspension/psychology , Humans , Mice , Synaptic Transmission
7.
Fish Physiol Biochem ; 48(1): 85-99, 2022 Feb.
Article En | MEDLINE | ID: mdl-34981327

We investigated whether dietary supplementation with Aurantiochytrium sp. meal, a DHA-rich source (docosahexaenoic acid, 22: 6 n-3), fed during long-term exposure to cold-suboptimal temperature (22 °C, P1), followed by short-term exposure to higher temperatures (28 °C, P2, and 33 °C, P3), would promote oxidative damage in Nile tilapia (Oreochromis niloticus). Two supplementation levels were tested: 1.0 g 100 g-1 (D1) and 4.0 g 100 g-1 (D4). A control diet, without the additive (D0, 0 g 100 g-1), and a positive control diet supplemented with cod liver oil (CLO) were also tested. The concentrations of DHA and total n-3 PUFAs in the CLO diet were similar to those found in diets D1 and D4, respectively. The parameters analyzed included hemoglobin (Hb), the antioxidant enzymes catalase, glutathione peroxidase, total glutathione, non-protein thiols, and the oxidative markers protein carbonyl and erythrocyte DNA damage. Nile tilapia did not present differences in Hb content, regardless of diet composition, but the temperature increase (P1 to P2) led to a higher Hb content. Likewise, the temperature increases promoted alterations in all antioxidant enzymes. The dietary supplementation with 1.0 g 100 g-1 Aurantiochytrium sp. meal after P1 caused minor DNA damage in Nile tilapia, demonstrating that the additive can safely be included in winter diets, despite its high DHA concentration.


Cichlids , Oxidative Stress , Temperature , Animal Feed/analysis , Animals , Antioxidants/metabolism , Cichlids/metabolism , Diet/veterinary , Dietary Supplements/analysis , Docosahexaenoic Acids/administration & dosage , Stramenopiles/chemistry
8.
Article En | MEDLINE | ID: mdl-34284103

The effect of increasing amounts (0%, 25%, 50%, 75%, and 100%) of dietary supplementation with an organic micromineral complex (Fe, Zn, Cu, Mn, and Se) on antioxidant defenses and mineral deposition in tissues of Nile tilapia juveniles was evaluated, where 100% supplementation represented the average adopted by the feed industry in Brazil. Fish (initial weight 23.93 ± 0.80 g) were fed until apparent satiation twice a day for 56 days. The maximum deposition of Fe and Zn in the hepatopancreas occurred in fish given approximately 50% supplementation, whereas the deposition of Mn and Se increased linearly with the inclusion of the complex. The activity of catalase and superoxide dismutase in the hepatopancreas decreased in fish fed the 50% dose, when compared to those not receiving mineral supplementation or those receiving higher doses. Glutathione peroxidase (GPx) activity in the hepatopancreas increased as the dietary Se concentration increased. However, the concentration of metallothionein in the hepatopancreas showed an inverse relationship to the increase in dietary supplementation of the organic mineral complex. There was no relationship between the doses of organic micromineral supplementation and the activities of GPx, reduced glutathione, non-protein thiols, or protein carbonylation. However, diets supplemented with 50% to 100% promoted greater GPx activity when compared to the 0% supplemented diet. Supplementation with intermediate doses of organic microminerals, approximately 50% of that used in commercial tilapia diets, promoted the homeostasis of metal metabolism, especially for Fe and Zn.


Animal Feed , Antioxidants/metabolism , Cichlids/physiology , Dietary Supplements , Metallothionein/metabolism , Animals , Antioxidants/chemistry , Brazil , Catalase/metabolism , Cichlids/metabolism , Diet , Glutathione , Glutathione Peroxidase/metabolism , Hepatopancreas/metabolism , Iron/chemistry , Male , Metallothionein/chemistry , Minerals/metabolism , Sulfhydryl Compounds/metabolism , Superoxide Dismutase/metabolism , Zinc/chemistry
9.
Mol Neurobiol ; 58(2): 735-749, 2021 Feb.
Article En | MEDLINE | ID: mdl-33011857

Methylglyoxal (MGO) is an endogenous toxin, mainly produced as a by-product of glycolysis that has been associated to aging, Alzheimer's disease, and inflammation. Cell culture studies reported that MGO could impair the glyoxalase, thioredoxin, and glutathione systems. Thus, we investigated the effect of in vivo MGO administration on these systems, but no major changes were observed in the glyoxalase, thioredoxin, and glutathione systems, as evaluated in the prefrontal cortex and the hippocampus of mice. A previous study from our group indicated that MGO administration produced learning/memory deficits and depression-like behavior. Confirming these findings, the tail suspension test indicated that MGO treatment for 7 days leads to depression-like behavior in three different mice strains. MGO treatment for 12 days induced working memory impairment, as evaluated in the Y maze spontaneous alternation test, which was paralleled by low dopamine and serotonin levels in the cerebral cortex. Increased DARPP32 Thr75/Thr34 phosphorylation ratio was observed, suggesting a suppression of phosphatase 1 inhibition, which may be involved in behavioral responses to MGO. Co-treatment with a dopamine/noradrenaline reuptake inhibitor (bupropion, 10 mg/kg, p.o.) reversed the depression-like behavior and working memory impairment and restored the serotonin and dopamine levels in the cerebral cortex. Overall, the cerebral cortex monoaminergic system appears to be a preferential target of MGO toxicity, a new potential therapeutic target that remains to be addressed.


Depression/physiopathology , Dopamine Uptake Inhibitors/pharmacology , Dopamine/deficiency , Memory, Short-Term , Norepinephrine/metabolism , Pyruvaldehyde/adverse effects , Animals , Bupropion/pharmacology , Dopamine/metabolism , Female , Glutathione/metabolism , Immobilization , Memory, Short-Term/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Motor Activity/drug effects , Phosphorylation/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pyruvaldehyde/administration & dosage , Serotonin/metabolism , Tyrosine 3-Monooxygenase/metabolism
10.
Acta Neurobiol Exp (Wars) ; 80(4): 364-374, 2020.
Article En | MEDLINE | ID: mdl-33350989

Memory impairment is a feature of several diseases and detrimental as aging population have increased worldwide. Sustained advanced glycation end products (AGEs) receptor (RAGE) activation triggers the production of reactive oxygen species and inflammatory response, leading to neuronal dysfunction and neurodegenerative disorders. Methylglyoxal (MGO) is the most relevant and reactive glycating agent in vivo, leading to the formation of AGEs. Here, we investigated the role of RAGE on the memory impairment induced by MGO. Swiss female mice were treated for 11 days with MGO, FPS­ZM1 (a high­affinity RAGE antagonist), or the combination of both. Locomotor activity was not impaired by the treatments, as evaluated by the open field and spontaneous alternation test. MGO treatment impaired short­ and long­term spatial memory in the object location task, caused deficits on the short­term aversive memory in the step­down inhibitory avoidance task, and decreased working memory performance as evaluated by the Y­maze spontaneous alternation test. FPS­ZM1 treatment abolished deficits on the short­term aversive memory and working memory, but was unable to prevent the impairment in short­term or long­term spatial memory. Since the addition of RAGE antagonist in co­treatment with MGO protected mice from the aversive and working memory deficits, AGEs generated by the MGO treatment would be involved in the memory impairment due to RAGE activation. Therefore, further studies are required to establish the involvement of RAGE in the MGO­induced memory impairment. Nevertheless, our results suggested FPS­ZM1 treatment as a promising new therapeutic strategy to prevent cognitive dysfunction caused by dicarbonyl stress, further investigation is required to confirm our findings.


Cognitive Dysfunction/drug therapy , Glycation End Products, Advanced/pharmacology , Memory Disorders/prevention & control , Neurons/drug effects , Pyruvaldehyde/pharmacology , Animals , Cell Survival/drug effects , Female , Memory Disorders/drug therapy , Mice
11.
Neurochem Res ; 45(12): 2868-2883, 2020 Dec.
Article En | MEDLINE | ID: mdl-32968860

Recent evidence suggests that young rodents submitted to high fructose (FRU) diet develop metabolic, and cognitive dysfunctions. However, it remains unclear whether these detrimental effects of FRU intake can also be observed in middle-aged mice. Nine months-old C57BL/6 female mice were fed with water (Control) or 10% FRU in drinking water during 12 weeks. After that, metabolic, and neurochemical alterations were evaluated, focusing on neurotransmitters, and antioxidant defenses. Behavioral parameters related to motor activity, memory, anxiety, and depression were also evaluated. Mice consuming FRU diet displayed increased water, and caloric intake, resulting in weight gain, which was partially compensated due to decreased food pellet intake. FRU fed animals displayed increased plasma glucose, and cholesterol levels, which was not observed in overnight-fasted animals. Superoxide dismutase (SOD), and catalase (CAT) activities were markedly decreased in the prefrontal cortex of animals receiving FRU diet, while glutathione peroxidase (GPx) slightly increased. Liver (lower GPx), striatum (higher SOD and lower CAT), and hippocampus (no changes) were less impacted. No changes were observed in glutathione reductase, and thioredoxin reductase activities, two ancillary enzymes for peroxide detoxification. FRU intake did not alter serotonin, dopamine, and norepinephrine levels in the hippocampus, prefrontal cortex, and striatum. No significant alterations were observed in working, and short-term spatial memory; and in anxiety- and depressive-like behaviors in animals treated with FRU. Increased locomotor activity was observed in FRU-fed middle-aged mice, as evaluated in the open field, elevated plus-maze, Y maze, and object location tasks. Overall, these results demonstrate that high FRU consumption can disturb antioxidant defenses, and increase locomotor activity in middle-aged mice, open the opportunity for further studies to address the underlying mechanisms related to these findings.


Catalase/metabolism , Fructose/pharmacology , Locomotion/drug effects , Superoxide Dismutase/metabolism , Animals , Brain/drug effects , Brain/metabolism , Elevated Plus Maze Test , Female , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL , Open Field Test/drug effects
12.
Food Res Int ; 136: 109441, 2020 10.
Article En | MEDLINE | ID: mdl-32846544

Processed meats are classified by the International Agency for Research on Cancer as category 1 because their consumption increase the incidence of colorectal and stomach cancers. Meat processing widely employs nitrite and sorbate as preservatives. When these preservatives are concomitantly used in non-compliant processes, they may react and produce the mutagen 2-methyl-1,4-dinitro-pyrrole (DNMP). This study aimed to evaluate the ability of different bacteria isolated from food matrices to biodegrade DNMP in in vitro reactions and in a processed meat model. A possible mechanism of biodegradation was also tested. In vitro experiments were performed in two steps. In the first one, only one strain out of 13 different species did not interact with DNMP. In the following step, an empirical conversion factor was calculated to assess the conversion of DNMP to 4-amino-2-methyl-1-nitro-pyrrole by the strains. The most efficient strains were Staphylococcus xylosus LYOCARNI SXH-01, Lactobacillus fermentum LB-UFSC 0017, and Lactobacillus casei LB-UFSC 0019, which yielded conversion factors of 0.62, 0.60, and 0.43, respectively. Thus, such strains were individually added to the processed meat model and completely degraded the DNMP. Moreover, S. xylosus degraded DNMP in less than 30 min. The enzymatic mechanism was evaluated using its cell-free extract. It showed that, in the aerobic system, reduction rates were 30.321 and 22.411 nmol/mg of protein/min using NADH and NADPH, respectively. A DNMP reductase was assigned to the extract and a potential presence of an oxygen insensitive nitroreductase type I B was considered. Thus, biotechnological processes may be an efficient strategy to eliminate the DNMP from meat products and to increase food safety.


Meat Products , Mutagens , Meat , Meat Products/analysis , Pyrroles , Staphylococcus
13.
Neurotoxicology ; 80: 144-154, 2020 09.
Article En | MEDLINE | ID: mdl-32738267

Exposure to fungicide ziram (zinc dimethyldithiocarbamate) has been associated with increased incidence of Parkinson's disease (PD). We recently demonstrated that the intranasal (i.n.) administration of sodium dimethyldithiocarbamate (NaDMDC, a more soluble salt than ziram) induces PD-like behavioral and neurochemical alterations in mice. We now investigated the putative neuroprotective effects of melatonin on behavioral dificits and neurochemical alterations induced by i.n. NaDMDC. Melatonin treatment (3, 10 or 30 mg/kg, i.p.) was given 1 h before NaDMDC administration (1 mg/nostril) during 4 consecutive days and we evaluated early (up to 7 days) and late (up to 35 days) NaDMDC-induced behavioral and neurochemical alterations. Melatonin treatment protected against early motor and general neurological impairments observed in the open field and neurological score of severity, respectively, and late deficits in rotarod test. Melatonin prevented the NaDMDC-induced alterations in the striatal tyrosine hydroxylase immunocontent. Melatonin also protected against increased levels of oxidative stress markers (4-hydroxynonenal and 3-nitrotyrosine) in the striatum, as well as the NaDMDC-induced increase of 4-hydroxynonenal and TNF, markers of oxidative stress and inflammation, respectively, in the olfactory bulb. These results further detail the mechanisms underlying NaDMDC toxicity and demonstrate the neuroprotective effects of melatonin against the neuronal damage induced by NaDMDC.


Brain/drug effects , Dopaminergic Neurons/drug effects , Melatonin/pharmacology , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/prevention & control , Administration, Intranasal , Animals , Behavior, Animal/drug effects , Brain/metabolism , Brain/physiopathology , Dimethyldithiocarbamate , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Male , Mice , Motor Activity/drug effects , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/physiopathology , Nitrosative Stress/drug effects , Oxidative Stress/drug effects
14.
Neurotox Res ; 38(3): 603-610, 2020 Oct.
Article En | MEDLINE | ID: mdl-32651842

Chlorpyrifos (CPF) is a neurotoxic organophosphorus (OP) insecticide widely used for agricultural purposes. CPF-mediated neurotoxicity is mainly associated with its anticholinesterase activity, which may lead to a cholinergic syndrome. CPF metabolism generates chlorpyrifos-oxon (CPF-O), which possesses higher anticholinesterase activity and, consequently, plays a major role in the cholinergic syndrome observed after CPF poisoning. Recent lines of evidence have also reported non-cholinergic endpoints of CPF- and CPF-O-induced neurotoxicities, but comparisons on the non-cholinergic toxic properties of CPF and CPF-O are lacking. In this study, we compared the non-cholinergic toxicities displayed by CPF and CPF-O in cultured neuronal cells, with a particular emphasis on their pro-oxidant properties. Using immortalized cells derived from mouse hippocampus (HT22 line, which does present detectable acetylcholinesterase activity), we observed that CPF-O was 5-fold more potent in decreasing cell viability compared with CPF. Atropine, a muscarinic acetylcholine receptor antagonist, protected against acetylcholine (ACh)-induced toxicity but failed to prevent the CPF- and CPF-O-induced cytotoxicities in HT22 cells. CPF or CPF-O exposures significantly decreased the levels of the antioxidant glutathione (GSH); this event preceded the significant decrease in cell viability. Pretreatment with N-acetylcysteine (NAC, a GSH precursor) protected against the cytotoxicity induced by both CPF and CPF-O. The present study indicates that GSH depletion is a non-cholinergic event involved in CPF and CPF-O toxicities. The study also shows that in addition of being a more potent AChE inhibitor, CPF-O is also a more potent pro-oxidant molecule when compared with CPF, highlighting the role of CPF metabolism (bioactivation to CPF-O) in the ensuing non-cholinergic toxicity.


Chlorpyrifos/analogs & derivatives , Glutathione/pharmacology , Neurons/drug effects , Neurotoxicity Syndromes/drug therapy , Acetylcholine/pharmacology , Acetylcholinesterase/metabolism , Animals , Atropine/pharmacology , Cell Survival/drug effects , Chlorpyrifos/pharmacology , Cholinesterase Inhibitors/pharmacology , Glutathione/metabolism
15.
Chem Biol Interact ; 328: 109195, 2020 Sep 01.
Article En | MEDLINE | ID: mdl-32707044

A previous study demonstrated that glutathione (GSH) produces specific antidepressant-like effect in the forced swimming test (FST), a predictive test of antidepressant activity. The present study investigated the involvement of multiple cellular targets implicated in the antidepressant-like effect of GSH in the FST. The antidepressant-like effect of GSH (300 nmol/site, icv) lasted up to 3 h when mice were submitted to FST. The central administration of oxidized GSH (GSSG, 3-300 nmol/site) did not alter the behavior of mice submitted to the FST. Furthermore, the combined treatment of sub-effective doses of GSH (100 nmol/site, icv) with a sub-effective dose of classical antidepressants (fluoxetine 10 mg/kg, and imipramine 5 mg/kg, ip) presented synergistic effect by decreasing the immobility time in the FST. The antidepressant-like effect of GSH was abolished by prazosin (1 mg/kg, ip, α1-adrenoceptor antagonist), baclofen (1 mg/kg, ip, GABAB receptor agonist), bicuculline (1 mg/kg, ip, GABAA receptor antagonist), l-arginine (750 mg/kg, ip, NO precursor), SNAP (25 µg/site, icv, NO donor), but not by yohimbine (1 mg/kg, ip, α2-adrenoceptor antagonist). The NMDA receptor antagonists, MK-801(0.001 mg/kg, ip) or GMP (0.5 mg/kg, ip), potentiated the effect of a sub-effective dose of GSH in the FST. These results suggest that the antidepressant-like effect induced by GSH is connected to the activation of α1 adrenergic and GABAA receptors, as well as the inhibition of GABAB and NMDA receptors and NO biosyntesis. We speculate that redox-mediated signaling on the extracelular portion of cell membrane receptors would be a common mechanism of action of GSH.


Antidepressive Agents/pharmacology , Glutathione/pharmacology , Molecular Targeted Therapy , Adrenergic Antagonists/pharmacology , Animals , Arginine/pharmacology , Drug Synergism , Female , Glutathione/administration & dosage , Hippocampus/drug effects , Hippocampus/metabolism , Immobilization , Male , Mice , Receptors, Adrenergic/metabolism , Receptors, GABA/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , S-Nitroso-N-Acetylpenicillamine/pharmacology , Swimming
16.
Mar Environ Res ; 154: 104870, 2020 Feb.
Article En | MEDLINE | ID: mdl-32056707

Glutathione (GSH) is a major cellular antioxidant molecule participating in several biological processes, including immune function. In this study, we investigated the importance of GSH to oysters Crassostrea gigas immune response. Oysters were treated with the GSH-synthesis inhibitor buthionine sulfoximine (BSO), and the function of immune cells and mortality were evaluated after a bacterial challenge with different Vibrio species. BSO caused a moderate decrease (20-40%) in GSH levels in the gills, digestive gland, and hemocytes. As expected, lower GSH decreased survival to peroxide exposure. Hemocyte function was preserved after BSO treatment, however, oysters became more susceptible to challenges with Vibrio anguillarum, V. alginolyticus, or V. harveyi, but not with V. parahaemolyticus and V. vulnificus, indicating a species-specific vulnerability. Our study indicates that in natural habitats or in mariculture farms, disturbances in GSH metabolism may pre-dispose oysters to bacterial infection, decreasing survival.


Crassostrea , Vibrio , Animals , Crassostrea/metabolism , Crassostrea/microbiology , Gills/metabolism , Gills/microbiology , Glutathione/metabolism , Hemocytes/metabolism , Hemocytes/microbiology , Vibrio/physiology
17.
Neurochem Int ; 132: 104585, 2020 01.
Article En | MEDLINE | ID: mdl-31678323

Thioredoxin interacting protein (TXNIP) binds to thioredoxin thereby limiting its activity, but it also promotes internalization of glucose transporters, participates in inflammasome activation, and controls autophagy. Published data and this work demonstrate that TXNIP responds to a number of apparently unrelated stresses, such as serum deprivation, pH change, and oxidative, osmotic and carbonyl stress. Interestingly, we noticed that hyperosmotic (NaCl) and carbonyl (methylglyoxal, MGO) stresses in HT22 neuronal cells produced a rapid loss of TXNIP (half-life ∼12 min), prompting us to search for possible mechanisms controlling this TXNIP loss, including pH change, serum deprivation, calcium metabolism and inhibition of the proteasome and other proteases, autophagy and MAPKs. None of these routes stopped the TXNIP loss induced by hyperosmotic and carbonyl stress. Besides transcriptional, translational and microRNA regulation, there is evidence indicating that mTOR and AMPK also control TXNIP expression. Indeed, AMPK-deficient mouse embryonic fibroblasts failed to respond to phenformin (AMPK activator) and compound C (AMPK inhibitor), while rapamycin induced a marked increase in TXNIP levels, confirming the known AMPK/mTOR control over TXNIP. However, the TXNIP loss induced by NaCl or MGO were observed even in AMPK deficient MEFs or after mTOR inhibition, indicating AMPK/mTOR does not participate in this rapid TXNIP loss. These results suggest that rapid TXNIP loss is a general and immediate response to stress that can improve energy availability and antioxidant protection, eventually culminating in better cell survival.


Carbon/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Neurons/metabolism , Osmotic Pressure/physiology , Oxidative Stress/physiology , Thioredoxins/antagonists & inhibitors , Thioredoxins/metabolism , Animals , Carrier Proteins/genetics , Cell Line, Transformed , Mice , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Thioredoxins/genetics , Time Factors
18.
Neurochem Res ; 45(2): 354-370, 2020 Feb.
Article En | MEDLINE | ID: mdl-31786717

Methylglyoxal (MGO) is a highly reactive dicarbonyl molecule that promotes the formation of advanced glycation end products (AGEs), which are believed to play a key role in a number of pathologies, such as diabetes, Alzheimer's disease, and inflammation. Here, Swiss mice were treated with MGO by intraperitoneal injection to investigate its effects on motor activity, mood, and cognition. Acute MGO treatment heavily decreased locomotor activity in the open field test at higher doses (80-200 mg/kg), an effect not observed at lower doses (10-50 mg/kg). Several alterations were observed 4 h after a single MGO injection (10-50 mg/kg): (a) plasma MGO levels were increased, (b) memory was impaired (object location task), (c) anxiolytic behavior was observed in the open field and marble burying test, and (d) depressive-like behavior was evidenced as evaluated by the tail suspension test. Biochemical alterations in the glutathione and glyoxalase systems were not observed 4 h after MGO treatment. Mice were also treated daily with MGO at 0, 10, 25 and 50 mg/kg for 11 days. From the 5th to the 11th day, several behavioral end points were evaluated, resulting in: (a) absence of motor impairment as evaluated in the open field, horizontal bars and pole test, (b) depressive-like behavior observed in the tail suspension test, and (c) cognitive impairments detected on working, short- and long-term memory when mice were tested in the Y-maze spontaneous alternation, object location and recognition tests, and step-down inhibitory avoidance task. An interesting finding was a marked decrease in dopamine levels in the prefrontal cortex of mice treated with 50 mg/kg MGO for 11 days, along with a ~ 25% decrease in the Glo1 content. The MGO-induced dopamine depletion in the prefrontal cortex may be related to the observed memory deficits and depressive-like behavior, an interesting topic to be further studied as a potentially novel route for MGO toxicity.


Anti-Anxiety Agents/toxicity , Depression/chemically induced , Dopamine/metabolism , Memory Disorders/chemically induced , Prefrontal Cortex/drug effects , Pyruvaldehyde/toxicity , Animals , Depression/metabolism , Female , Lactoylglutathione Lyase/metabolism , Locomotion/drug effects , Memory/drug effects , Memory Disorders/metabolism , Mice , Prefrontal Cortex/metabolism
19.
Ecotoxicol Environ Saf ; 182: 109411, 2019 Oct 30.
Article En | MEDLINE | ID: mdl-31299475

The urban growth has increased sanitary sewage discharges in coastal ecosystems, negatively affecting the aquatic biota. Mangroves, one of the most human-affected coastal biomes, are areas for reproduction and nursing of several species. In order to evaluate the effects of sanitary sewage effluents in mangrove species, this study assessed the hepatic transcriptional responses of guppy fish Poecilia vivipara exposed to sanitary sewage 33% (v:v), using suppressive subtraction hybridization (SSH), high throughput sequencing of RNA (Ion-proton) and quantification of transcript levels by qPCR of some identified genes in fish kept in a sewage-contaminated environment. Genes identified are related predominantly to xenobiotic biotransformation, immune system and sexual differentiation. The qPCR results confirmed the induction of cytochrome P450 1A (CYP1A), glutathione S transferase A-like (GST A-like) methyltransferase (MET) and UDP glycosyltransferase 1A (UDPGT1A), and repression of complement component C3 (C3), doublesex and mab-3 related transcription factor 1 (DMRT1), and transferrin (TF) in the laboratory experiment. In the field exposure, the transcript levels of CYP1A, DMRT1, MET, GST A-like and UDPGT1A were higher in fishes exposed at the contaminated sites compared to the reference site. Chemical analysis in fish from the laboratory and in situ experiments, and surface sediment from the sewage-contaminated sites revealed relevant levels of polycyclic aromatic hydrocarbons (PAHs), polychlorinated biphenyl (PCBs) and linear alkylbenzenes (LABs). These data reinforce the use of P. vivipara as a sentinel for monitoring environmental contamination in coastal regions.


Environmental Monitoring/methods , Liver/drug effects , Poecilia/genetics , Sewage/chemistry , Transcription, Genetic/drug effects , Water Pollutants, Chemical/toxicity , Animals , Biotransformation , Estuaries , Liver/metabolism , Models, Theoretical , Poecilia/metabolism , Water Pollutants, Chemical/metabolism , Xenobiotics/metabolism
20.
Oxid Med Cell Longev ; 2019: 2715810, 2019.
Article En | MEDLINE | ID: mdl-31049129

BACKGROUND: Hyperosmotic stress is an important pathophysiologic condition in diabetes, severe trauma, dehydration, infection, and ischemia. Furthermore, brain neuronal cells face hyperosmotic stress in ageing and Alzheimer's disease. Despite the enormous importance of knowing the homeostatic mechanisms underlying the responses of nerve cells to hyperosmotic stress, this topic has been underrepresented in the literature. Recent evidence points to autophagy induction as a hallmark of hyperosmotic stress, which has been proposed to be controlled by mTOR inhibition as a consequence of AMPK activation. We previously showed that methylglyoxal induced a decrease in the antioxidant proteins thioredoxin 1 (Trx1) and glyoxalase 2 (Glo2), which was mediated by AMPK-dependent autophagy. Thus, we hypothesized that hyperosmotic stress would have the same effect. METHODS: HT22 hippocampal nerve cells were treated with NaCl (37, 75, or 150 mM), and the activation of the AMPK/mTOR pathway was investigated, as well as the levels of Trx1 and Glo2. To determine if autophagy was involved, the inhibitors bafilomycin (Baf) and chloroquine (CQ), as well as ATG5 siRNA, were used. To test for AMPK involvement, AMPK-deficient mouse embryonic fibroblasts (MEFs) were used. RESULTS: Hyperosmotic stress induced a clear increase in autophagy, which was demonstrated by a decrease in p62 and an increase in LC3 lipidation. AMPK phosphorylation, linked to a decrease in mTOR and S6 ribosomal protein phosphorylation, was also observed. Deletion of AMPK in MEFs did not prevent autophagy induction by hyperosmotic stress, as detected by decreased p62 and increased LC3 II, or mTOR inhibition, inferred by decreased phosphorylation of P70 S6 kinase and S6 ribosomal protein. These data indicating that AMPK was not involved in autophagy activation by hyperosmotic stress were supported by a decrease in pS555-ULK1, an AMPK phosphorylation site. Trx1 and Glo2 levels were decreased at 6 and 18 h after treatment with 150 mM NaCl. However, this decrease in Trx1 and Glo2 in HT22 cells was not prevented by autophagy inhibition by Baf, CQ, or ATG5 siRNA. AMPK-deficient MEFs under hyperosmotic stress presented the same Trx1 and Glo2 decrease as wild-type cells. CONCLUSION: Hyperosmotic stress induced AMPK activation, but this was not responsible for its effects on mTOR activity or autophagy induction. Moreover, the decrease in Trx1 and Glo2 induced by hyperosmotic stress was independent of both autophagy and AMPK activation.


AMP-Activated Protein Kinases/metabolism , Autophagy , Neurons/metabolism , Osmotic Pressure , Signal Transduction , Thiolester Hydrolases/metabolism , Thioredoxins/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Line, Transformed , Enzyme Activation , Mice , Neurons/cytology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Thiolester Hydrolases/genetics , Thioredoxins/genetics
...