Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Antioxidants (Basel) ; 12(10)2023 Oct 03.
Article En | MEDLINE | ID: mdl-37891904

Concerns have been raised regarding the potential adverse health effects of the ubiquitous herbicide glyphosate. Here, we investigated long-term effects of developmental exposure to a glyphosate-based herbicide (GBH) by analyzing serum melatonin levels and cellular changes in the striatum of adult male rats (90 days old). Pregnant and lactating rats were exposed to 3% GBH (0.36% glyphosate) through drinking water from gestational day 5 to postnatal day 15. The offspring showed reduced serum melatonin levels (43%) at the adult age compared with the control group. The perinatal exposure to GBH also induced long-term oxidative stress-related changes in the striatum demonstrated by increased lipid peroxidation (45%) and DNA/RNA oxidation (39%) together with increased protein levels of the antioxidant enzymes, superoxide dismutase (SOD1, 24%), glutamate-cysteine ligase (GCLC, 58%), and glutathione peroxidase 1 (GPx1, 31%). Moreover, perinatal GBH exposure significantly increased the total number of neurons (20%) and tyrosine hydroxylase (TH)-positive neurons (38%) in the adult striatum. Mechanistic in vitro studies with primary rat pinealocytes exposed to 50 µM glyphosate demonstrated a decreased melatonin secretion partially through activation of metabotropic glutamate receptor 3 (mGluR3), while higher glyphosate levels (100 or 500 µM) also reduced the pinealocyte viability. Since decreased levels of the important antioxidant and neuroprotector melatonin have been associated with an increased risk of developing neurodegenerative disorders, this demonstrates the need to consider the melatonin hormone system as a central endocrine-related target of glyphosate and other environmental contaminants.

2.
Toxicol Appl Pharmacol ; 454: 116245, 2022 11 01.
Article En | MEDLINE | ID: mdl-36116562

The present study investigated the effects of perinatal exposure to glyphosate-based herbicide (GBH) in offspring's liver. Pregnant Wistar rats were exposed to GBH (70 mg glyphosate/Kg body weight/day) in drinking water from gestation day 5 to postnatal day 15. The perinatal exposure to GBH increased 45Ca2+ influx in offspring's liver. Pharmacological tools indicated a role played by oxidative stress, phospholipase C (PLC) and Akt pathways, as well as voltage-dependent Ca2+ channel modulation on GBH-induced Ca2+ influx in offspring's liver. In addition, changes in the enzymatic antioxidant defense system, decreased GSH content, lipid peroxidation and protein carbonylation suggest a connection between GBH-induced hepatotoxic mechanism and redox imbalance. The perinatal exposure to GBH also increased the enzymatic activities of transaminases and gamma-glutamyl transferase in offspring's liver and blood, suggesting a pesticide-induced liver injury. Moreover, we detected increased iron levels in liver, blood and bone marrow of GBH-exposed rats, which were accompanied by increased transferrin saturation and decreased transferrin levels in blood. The levels of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) were increased in the liver of rats perinatally exposed to GBH, which were associated with. Increased phospho-p65NFκB immunocontent. Therefore, we propose that excessive amounts of iron in offspring's liver, blood and bone marrow induced by perinatal exposure to GBH may account for iron-driven hepatotoxicity, which was associated with Ca2+ influx, oxidative damage and inflammation. Further studies will clarify whether these events can ultimately impact on liver function.


Drinking Water , Herbicides , Liver Diseases , Pesticides , Animals , Antioxidants , Female , Glycine/analogs & derivatives , Herbicides/toxicity , Interleukin-6 , Iron , Pregnancy , Proto-Oncogene Proteins c-akt , Rats , Rats, Wistar , Transaminases , Transferrins , Tumor Necrosis Factor-alpha , Type C Phospholipases , Glyphosate
3.
Sci Total Environ ; 808: 151945, 2022 Feb 20.
Article En | MEDLINE | ID: mdl-34843762

Environmental contaminants including long-chain per- and polyfluoroalkyl substances (PFAS) have been linked to cancer, which is a central cause of mortality in humans and many wildlife species. Today shorter-chain PFAS are extensively used as replacement compounds and commonly found in the environment. Mechanistic studies are important for a better understanding of their toxicological potential and possible role in cancer etiology. Here, we treated normal human breast epithelial cells (MCF-10A) with 500 pM to 500 µM of perfluorohexane sulfonate (PFHxS), undecafluorohexanoic acid (PFHxA), hexafluoropropylene oxide-dimer acid (GenX), perfluoro 3,6 dioxaoctanoic acid (PFO2OA), heptafluorobutyric acid (HFBA) and perfluorobutanesulfonic acid (PFBS) for 72 h to investigate potential effects on cell proliferation and neoplastic transformation. PFHxA, GenX, PFO2OA, HFBA and PFBS induced no alterations compared to controls at any of the concentrations tested. Exposure to 100 µM PFHxS on the other hand was shown to affect important regulatory cell-cycle proteins (cyclin D1, CDK6, p27, p53 and ERK) and induced cell proliferation, at least in part through activation of the constitutive androstane receptor (CAR) and the peroxisome proliferator-activated receptor alpha (PPARα). PFHxS also altered histone modifications and induced cell malignance by reducing the levels of adhesion proteins (E-cadherin and ß-integrin) and promoting cell migration and invasion. These results demonstrate that five out of six alternative PFAS tested are clearly less harmful to MCF-10A cells than previously studied PFOS and PFOA, but raise concerns about PFHxS that also has been associated with breast cancer in epidemiological studies.


Alkanesulfonic Acids , Environmental Pollutants , Fluorocarbons , Alkanesulfonates , Alkanesulfonic Acids/toxicity , Carcinogenesis , Constitutive Androstane Receptor , Fluorocarbons/analysis , Fluorocarbons/toxicity , Humans
4.
Toxicology ; 461: 152922, 2021 09.
Article En | MEDLINE | ID: mdl-34474092

Glyphosate, the most used herbicide worldwide, has been suggested to induce neurotoxicity and behavioral changes in rats after developmental exposure. Studies of human glyphosate intoxication have reported adverse effects on the nervous system, particularly in substantia nigra (SN). Here we used matrix-assisted laser desorption ionization (MALDI) imaging mass spectrometry (IMS) to study persistent changes in peptide expression in the SN of 90-day-old adult male Wistar rats. The animals were perinatally exposed to 3 % GBH (glyphosate-based herbicide) in drinking water (corresponding to 0.36 % of glyphosate) starting at gestational day 5 and continued up to postnatal day 15 (PND15). Peptides are present in the central nervous system before birth and play a critical role in the development and survival of neurons, therefore, observed neuropeptide changes could provide better understanding of the GBH-induced long term effects on SN. The results revealed 188 significantly altered mass peaks in SN of animals perinatally exposed to GBH. A significant reduction of the peak intensity (P < 0.05) of several peptides from the opioid-related dynorphin family such as dynorphin B (57 %), alpha-neoendorphin (50 %), and its endogenous metabolite des-tyrosine alpha-neoendorphin (39 %) was detected in the GBH group. Immunohistochemical analysis confirmed a decreased dynorphin expression and showed a reduction of the total area of dynorphin immunoreactive fibers in the SN of the GBH group. In addition, a small reduction of dynorphin immunoreactivity associated with non-neuronal cells was seen in the hilus of the hippocampal dentate gyrus. Perinatal exposure to GBH also induced an increase in the number of nestin-positive cells in the subgranular zone of the dentate gyrus. In conclusion, the results demonstrate long-term changes in the adult male rat SN and hippocampus following a perinatal GBH exposure suggesting that this glyphosate-based formulation may perturb critical neurodevelopmental processes.


Dynorphins/metabolism , Glycine/analogs & derivatives , Herbicides/toxicity , Neurotoxicity Syndromes/etiology , Animals , Brain/drug effects , Brain/pathology , Female , Glycine/administration & dosage , Glycine/toxicity , Herbicides/administration & dosage , Hippocampus/drug effects , Hippocampus/pathology , Male , Neurotoxicity Syndromes/pathology , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Wistar , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Glyphosate
5.
Neurotoxicology ; 85: 121-132, 2021 07.
Article En | MEDLINE | ID: mdl-34048864

Paraquat (1,1'-dimethyl-4,4'-bipyridinium dichloride; PQ) is a widely used herbicide in Brazilian crops, despite its banishment in many other countries. The present study investigated the effects of repeated dose of PQ on glutamate system, energy metabolism and redox parameters in the hippocampus of prepubertal rats. Twenty-two-day-old rats received daily intraperitoneal injections of PQ (10 mg/Kg) during 5 consecutive days and the effects of the pesticide were assessed 24 h after the last injection. The PQ exposure provoked cytotoxicity associated to decreased cell viability and increased glutamate excitotoxicity, as demonstrated by decreased 14C-glutamate uptake and increased 45Ca2+ uptake. Downregulated glutamine synthetase (GS) activity, further supports disrupted glutamate metabolism compromising the glutamate-glutamine cycle. Downregulated 14C-2-Deoxy-D-glucose indicates energy failure and upregulated lactate dehydrogenase (LDH) suggests the relevance of lactate as energy fuel. Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) upregulation suggest Krebs cycle replenishment and piruvate production. In addition, PQ disturbed the redox status inducing lipid peroxidation, evaluated by increased TBARS and imbalanced antioxidant system. Downregulated glutathione reductase (GR), gamma-glutamyltransferase (GGT), glutathione-S-transferase (GST) and glucose-6-P-dehydrogenase (G6PD) activities together with upregulated superoxide dismutase (SOD) and catalase activities corroborate the oxidative imbalance. The mechanisms underlying PQ-induced neurotoxicity involves the modulation of GSK-3ß, NF-κB and NMDA receptors. These neurochemical and oxidative events observed may contribute to neuroinflammation and neurotoxic effects of PQ on hippocampal cells.


Energy Metabolism/drug effects , Glutamic Acid/metabolism , Herbicides/toxicity , Hippocampus/metabolism , Paraquat/toxicity , Sexual Maturation/drug effects , Animals , Cell Survival/drug effects , Cell Survival/physiology , Energy Metabolism/physiology , Hippocampus/drug effects , Male , Organ Culture Techniques , Oxidation-Reduction/drug effects , Rats , Rats, Wistar , Sexual Maturation/physiology
6.
Cell Death Dis ; 11(10): 910, 2020 10 24.
Article En | MEDLINE | ID: mdl-33099583

Developmental exposure to the environmental neurotoxin ß-N-methylamino-L-alanine (BMAA), a proposed risk factor for neurodegenerative disease, can induce long-term cognitive impairments and neurodegeneration in rats. While rodent studies have demonstrated a low transfer of BMAA to the adult brain, this toxin is capable to cross the placental barrier and accumulate in the fetal brain. Here, we investigated the differential susceptibility of primary neuronal cells and neural stem cells from fetal rat hippocampus to BMAA toxicity. Exposure to 250 µM BMAA induced cell death in neural stem cells through caspase-independent apoptosis, while the proliferation of primary neurons was reduced only at 3 mM BMAA. At the lowest concentrations tested (50 and 100 µM), BMAA disrupted neural stem cell differentiation and impaired neurite development in neural stem cell-derived neurons (e.g., reduced neurite length, the number of processes and branches per cell). BMAA induced no alterations of the neurite outgrowth in primary neurons. This demonstrates that neural stem cells are more susceptible to BMAA exposure than primary neurons. Importantly, the changes induced by BMAA in neural stem cells were mitotically inherited to daughter cells. The persistent nature of the BMAA-induced effects may be related to epigenetic alterations that interfere with the neural stem cell programming, as BMAA exposure reduced the global DNA methylation in the cells. These findings provide mechanistic understanding of how early-life exposure to BMAA may lead to adverse long-term consequences, and potentially predispose for neurodevelopmental disorders or neurodegenerative disease later in life.


Amino Acids, Diamino/toxicity , DNA Methylation/drug effects , Hippocampus/drug effects , Nervous System Diseases/pathology , Neural Stem Cells/drug effects , Neurites/drug effects , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cyanobacteria Toxins , Female , Hippocampus/cytology , Hippocampus/metabolism , Nervous System Diseases/chemically induced , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurites/metabolism , Pregnancy , Rats , Rats, Wistar
8.
Arch Toxicol ; 94(11): 3893-3906, 2020 11.
Article En | MEDLINE | ID: mdl-32700164

Gene-environment interactions are involved in the development of breast cancer, the tumor type that accounts for the majority of the cancer-related deaths among women. Here, we demonstrate that exposure to PFOS (10 µM) and PFOA (100 µM)-two contaminants ubiquitously found in human blood-for 72 h induced breast epithelial cell (MCF-10A cell line) proliferation and alteration of regulatory cell-cycle proteins (cyclin D1, CDK6, p21, p53, p27, ERK 1/2 and p38) that persisted after a multitude of cell divisions. The contaminants also promoted cell migration and invasion by reducing the levels of E-cadherin, occludin and ß-integrin in the unexposed daughter cells. The compounds further induced an increase in global DNA methylation and differentially altered histone modifications, epigenetic mechanisms implicated in tumorigenesis. This mechanistic evidence for PFOS- and PFOA-induced malignant transformation of human breast cells supports a role of these abundant contaminants in the development and progression of breast cancer. Increased knowledge of contaminant-induced effects and their contribution to breast tumorigenesis is important for a better understanding of gene-environment interactions in the etiology of breast cancer.


Alkanesulfonic Acids/toxicity , Breast Neoplasms/chemically induced , Caprylates/toxicity , Carcinogenesis/chemically induced , Cell Cycle Proteins/metabolism , DNA Methylation/drug effects , Epigenesis, Genetic/drug effects , Fluorocarbons/toxicity , Breast Neoplasms/genetics , Carcinogenesis/genetics , Cell Cycle/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Female , Humans
9.
Mol Cell Endocrinol ; 470: 281-294, 2018 07 15.
Article En | MEDLINE | ID: mdl-29155306

In the present study we provide evidence that 3,3',5'-triiodothyronine (reverse T3, rT3) restores neurochemical parameters induced by congenital hypothyroidism in rat hippocampus. Congenital hypothyroidism was induced by adding 0.05% propylthiouracil in the drinking water from gestation day 8 and continually up to lactation day 15. In the in vivo rT3 exposure, hypothyroid 12-day old pups were daily injected with rT3 (50 ng/kg body weight) or saline until day 14. In the ex vivo rT3 treatment, hippocampal slices from 15-day-old hypothyroid pups were incubated for 30 min with or without rT3 (1 nM). We found that ex vivo and/or in vivo exposure to rT3 failed in restoring the decreased 14C-glutamate uptake; however, restored the phosphorylation of glial fibrillary acidic protein (GFAP), 45Ca2+ influx, aspartate transaminase (AST), glutamine synthetase (GS) and gamma-glutamate transferase (GGT) activities, as well as glutathione (GSH) levels in hypothyroid hippocampus. In addition, rT3 improved 14C-2-deoxy-D-glucose uptake and lactate dehydrogenase (LDH) activity. Receptor agonists/antagonists (RGD peptide and AP-5), kinase inhibitors of p38MAPK, ERK1/2, CaMKII, PKA (SB239063, PD98059, KN93 and H89, respectively), L-type voltage-dependent calcium channel blocker (nifedipine) and intracellular calcium chelator (BAPTA-AM) were used to determine the mechanisms of the nongenomic rT3 action on GGT activity. Using molecular docking analysis, we found rT3 interaction with αvß3 integrin receptors, nongenomically activating signaling pathways (PKA, CaMKII, p38MAPK) that restored GGT activity. We provide evidence that rT3 is an active TH metabolite and our results represent an important contribution to elucidate the nonclassical mechanism of action of this metabolite in hypothyroidism.


Hippocampus/enzymology , Hypothyroidism/enzymology , Integrin alphaVbeta3/metabolism , Signal Transduction , Triiodothyronine, Reverse/pharmacology , Animals , Biological Transport/drug effects , Calcium/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glucose/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamic Acid/metabolism , Glutathione/metabolism , Homeostasis/drug effects , Hypothyroidism/pathology , L-Lactate Dehydrogenase/metabolism , Models, Biological , Molecular Docking Simulation , Phosphorylation/drug effects , Rats, Wistar , Receptors, Glutamate/metabolism , Signal Transduction/drug effects , Transaminases/metabolism
10.
Toxicology ; 387: 67-80, 2017 07 15.
Article En | MEDLINE | ID: mdl-28627408

We have previously demonstrated that maternal exposure to glyphosate-based herbicide (GBH) leads to glutamate excitotoxicity in 15-day-old rat hippocampus. The present study was conducted in order to investigate the effects of subchronic exposure to GBH on some neurochemical and behavioral parameters in immature and adult offspring. Rats were exposed to 1% GBH in drinking water (corresponding to 0.36% of glyphosate) from gestational day 5 until postnatal day (PND)-15 or PND60. Results showed that GBH exposure during both prenatal and postnatal periods causes oxidative stress, affects cholinergic and glutamatergic neurotransmission in offspring hippocampus from immature and adult rats. The subchronic exposure to the pesticide decreased L-[14C]-glutamate uptake and increased 45Ca2+ influx in 60-day-old rat hippocampus, suggesting a persistent glutamate excitotoxicity from developmental period (PND15) to adulthood (PND60). Moreover, GBH exposure alters the serum levels of the astrocytic protein S100B. The effects of GBH exposure were associated with oxidative stress and depressive-like behavior in offspring on PND60, as demonstrated by the prolonged immobility time and decreased time of climbing observed in forced swimming test. The mechanisms underlying the GBH-induced neurotoxicity involve the NMDA receptor activation, impairment of cholinergic transmission, astrocyte dysfunction, ERK1/2 overactivation, decreased p65 NF-κB phosphorylation, which are associated with oxidative stress and glutamate excitotoxicity. These neurochemical events may contribute, at least in part, to the depressive-like behavior observed in adult offspring.


Behavior, Animal/drug effects , Depression/chemically induced , Glutamic Acid/metabolism , Glycine/analogs & derivatives , Herbicides/toxicity , Hippocampus/drug effects , Neurotoxicity Syndromes/etiology , Oxidative Stress/drug effects , Prenatal Exposure Delayed Effects , Acetylcholinesterase/metabolism , Age Factors , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Binding Sites , Cholinergic Fibers/drug effects , Cholinergic Fibers/metabolism , Depression/metabolism , Depression/physiopathology , Depression/psychology , Female , GPI-Linked Proteins/metabolism , Gestational Age , Glutamic Acid/chemistry , Glycine/chemistry , Glycine/metabolism , Glycine/toxicity , Herbicides/chemistry , Herbicides/metabolism , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Mitogen-Activated Protein Kinases/metabolism , Molecular Docking Simulation , Molecular Structure , Motor Activity/drug effects , NF-kappa B/metabolism , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/physiopathology , Neurotoxicity Syndromes/psychology , Pregnancy , Protein Binding , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/metabolism , S100 Calcium Binding Protein beta Subunit/metabolism , Structure-Activity Relationship , Synaptic Transmission/drug effects , Glyphosate
11.
Alcohol Clin Exp Res ; 40(1): 52-61, 2016 Jan.
Article En | MEDLINE | ID: mdl-26727523

BACKGROUND: Alcohol abuse during pregnancy leads to intellectual disability and morphological defects in the offspring. The aim of this study was to determine the effect of chronic maternal ethanol (EtOH) consumption during pregnancy and lactation on glutamatergic transmission regulation, energy deficit, and oxidative stress in the hippocampus of the offspring. METHODS: EtOH was administered to dams in drinking water at increasing doses (2 to 20%) from the gestation day 5 to lactation day 21. EtOH and tap water intake by treated and control groups, respectively, were measured daily. RESULTS: Results showed that EtOH exposure does not affect fluid intake over the course of pregnancy and lactation. The toxicity of maternal exposure to EtOH was demonstrated by decreased offspring body weight at experimental age, on postnatal day 21. Moreover, maternal EtOH exposure decreased (45) Ca(2+) influx in the offspring's hippocampus. Corroborating this finding, EtOH increased both Na(+) -dependent and Na(+) -independent glial [(14) C]-glutamate uptake in hippocampus of immature rats. Also, maternal EtOH exposure decreased glutamine synthetase activity and induced aspartate aminotransferase enzymatic activity, suggesting that in EtOH-exposed offspring hippocampus, glutamate is preferentially used as a fuel in tricarboxylic acid cycle instead of being converted into glutamine. In addition, EtOH exposure decreased [U-14C]-2-deoxy-D-glucose uptake in offspring hippocampus. CONCLUSIONS: The decline in glucose transport coincided with increased lactate dehydrogenase activity, suggesting an adaptative response in EtOH-exposed offspring hippocampus, using lactate as an alternative fuel. These events were associated with oxidative damage, as demonstrated by changes in the enzymatic antioxidant defense system and lipid peroxidation. Taken together, the results demonstrate that maternal exposure to EtOH during pregnancy and lactation impairs glutamatergic transmission, as well as inducing oxidative stress and energy deficit in immature rat hippocampus.


Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Glutamic Acid/metabolism , Glutamine/metabolism , Hippocampus/drug effects , Maternal Exposure , Neuroglia/drug effects , Oxidative Stress/drug effects , Prenatal Exposure Delayed Effects/metabolism , Animals , Animals, Newborn , Calcium/metabolism , Carbon Radioisotopes , Female , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Lactation , Neuroglia/metabolism , Pregnancy , Radionuclide Imaging , Rats , Rats, Wistar , Sodium/metabolism
12.
Toxicology ; 320: 34-45, 2014 Jun 05.
Article En | MEDLINE | ID: mdl-24636977

Previous studies demonstrate that glyphosate exposure is associated with oxidative damage and neurotoxicity. Therefore, the mechanism of glyphosate-induced neurotoxic effects needs to be determined. The aim of this study was to investigate whether Roundup(®) (a glyphosate-based herbicide) leads to neurotoxicity in hippocampus of immature rats following acute (30min) and chronic (pregnancy and lactation) pesticide exposure. Maternal exposure to pesticide was undertaken by treating dams orally with 1% Roundup(®) (0.38% glyphosate) during pregnancy and lactation (till 15-day-old). Hippocampal slices from 15 day old rats were acutely exposed to Roundup(®) (0.00005-0.1%) during 30min and experiments were carried out to determine whether glyphosate affects (45)Ca(2+) influx and cell viability. Moreover, we investigated the pesticide effects on oxidative stress parameters, (14)C-α-methyl-amino-isobutyric acid ((14)C-MeAIB) accumulation, as well as glutamate uptake, release and metabolism. Results showed that acute exposure to Roundup(®) (30min) increases (45)Ca(2+) influx by activating NMDA receptors and voltage-dependent Ca(2+) channels, leading to oxidative stress and neural cell death. The mechanisms underlying Roundup(®)-induced neurotoxicity also involve the activation of CaMKII and ERK. Moreover, acute exposure to Roundup(®) increased (3)H-glutamate released into the synaptic cleft, decreased GSH content and increased the lipoperoxidation, characterizing excitotoxicity and oxidative damage. We also observed that both acute and chronic exposure to Roundup(®) decreased (3)H-glutamate uptake and metabolism, while induced (45)Ca(2+) uptake and (14)C-MeAIB accumulation in immature rat hippocampus. Taken together, these results demonstrated that Roundup(®) might lead to excessive extracellular glutamate levels and consequently to glutamate excitotoxicity and oxidative stress in rat hippocampus.


Glycine/analogs & derivatives , Herbicides/toxicity , Hippocampus/drug effects , Neurotoxicity Syndromes/etiology , Prenatal Exposure Delayed Effects/physiopathology , Animals , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glutamic Acid/metabolism , Glycine/administration & dosage , Glycine/toxicity , Herbicides/administration & dosage , Hippocampus/pathology , Lactation/metabolism , Male , Maternal Exposure/adverse effects , Neurotoxicity Syndromes/physiopathology , Oxidative Stress/drug effects , Pregnancy , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Glyphosate
13.
Free Radic Biol Med ; 65: 335-346, 2013 Dec.
Article En | MEDLINE | ID: mdl-23820267

Glyphosate is the primary active constituent of the commercial pesticide Roundup. The present results show that acute Roundup exposure at low doses (36 ppm, 0.036 g/L) for 30 min induces oxidative stress and activates multiple stress-response pathways leading to Sertoli cell death in prepubertal rat testis. The pesticide increased intracellular Ca(2+) concentration by opening L-type voltage-dependent Ca(2+) channels as well as endoplasmic reticulum IP3 and ryanodine receptors, leading to Ca(2+) overload within the cells, which set off oxidative stress and necrotic cell death. Similarly, 30 min incubation of testis with glyphosate alone (36 ppm) also increased (45)Ca(2+) uptake. These events were prevented by the antioxidants Trolox and ascorbic acid. Activated protein kinase C, phosphatidylinositol 3-kinase, and the mitogen-activated protein kinases such as ERK1/2 and p38MAPK play a role in eliciting Ca(2+) influx and cell death. Roundup decreased the levels of reduced glutathione (GSH) and increased the amounts of thiobarbituric acid-reactive species (TBARS) and protein carbonyls. Also, exposure to glyphosate-Roundup stimulated the activity of glutathione peroxidase, glutathione reductase, glutathione S-transferase, γ-glutamyltransferase, catalase, superoxide dismutase, and glucose-6-phosphate dehydrogenase, supporting downregulated GSH levels. Glyphosate has been described as an endocrine disruptor affecting the male reproductive system; however, the molecular basis of its toxicity remains to be clarified. We propose that Roundup toxicity, implicated in Ca(2+) overload, cell signaling misregulation, stress response of the endoplasmic reticulum, and/or depleted antioxidant defenses, could contribute to Sertoli cell disruption in spermatogenesis that could have an impact on male fertility.


Glycine/analogs & derivatives , Herbicides/toxicity , Sertoli Cells/drug effects , Testis/drug effects , Animals , Blotting, Western , Calcium/metabolism , Glycine/toxicity , Male , Necrosis/chemically induced , Oxidative Stress/physiology , Rats , Rats, Wistar , Sertoli Cells/pathology , Testis/pathology , Glyphosate
14.
Mol Cell Endocrinol ; 375(1-2): 14-26, 2013 Aug 15.
Article En | MEDLINE | ID: mdl-23693027

Congenital hypothyroidism is associated with delay in cell migration and proliferation in brain tissue, impairment of synapse formation, misregulation of neurotransmitters, hypomyelination and mental retardation. However, the mechanisms underlying the neuropsychological deficits observed in congenital hypothyroidism are not completely understood. In the present study we proposed a mechanism by which hypothyroidism leads to hippocampal neurotoxicity. Congenital hypothyroidism induces c-Jun-N-terminal kinase (JNK) pathway activation leading to hyperphosphorylation of the glial fibrillary acidic protein (GFAP), vimentin and neurofilament subunits from hippocampal astrocytes and neurons, respectively. Moreover, hyperphosphorylation of the cytoskeletal proteins was not reversed by T3 and poorly reversed by T4. In addition, congenital hypothyroidism is associated with downregulation of astrocyte glutamate transporters (GLAST and GLT-1) leading to decreased glutamate uptake and subsequent influx of Ca(2+) through N-methyl-D-aspartate (NMDA) receptors. The Na(+)-coupled (14)C-α-methyl-amino-isobutyric acid ((14)C-MeAIB) accumulation into hippocampal cells also might cause an increase in the intracellular Ca(2+) concentration by opening voltage-dependent calcium channels (VDCC). The excessive influx of Ca(2+) through NMDA receptors and VDCCs might lead to an overload of Ca(2+) within the cells, which set off glutamate excitotoxicity and oxidative stress. The inhibited acetylcholinesterase (AChE) activity might also induce Ca(2+) influx. The inhibited glucose-6-phosphate dehydrogenase (G6PD) and gamma-glutamyl transferase (GGT) activities, associated with altered glutamate and neutral amino acids uptake could somehow affect the GSH turnover, the antioxidant defense system, as well as the glutamate-glutamine cycle. Reduced levels of S100B and glial fibrillary acidic protein (GFAP) take part of the hypothyroid condition, suggesting a compromised astroglial/neuronal neurometabolic coupling which is probably related to the neurotoxic damage in hypothyroid brain.


Acetylcholinesterase/metabolism , Congenital Hypothyroidism/enzymology , Hippocampus/enzymology , Oxidative Stress , Animals , Astrocytes/metabolism , Cell Count , Congenital Hypothyroidism/pathology , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 2/metabolism , GPI-Linked Proteins/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Hippocampus/growth & development , Hippocampus/pathology , Male , Neurons/metabolism , Rats , Rats, Wistar , S100 Calcium Binding Protein beta Subunit/metabolism , Thyroid Hormones/blood
...