Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 67
2.
Mol Cell Endocrinol ; 478: 62-76, 2018 12 15.
Article En | MEDLINE | ID: mdl-30031104

In this study, we used an experimental model of congenital hypothyroidism to show that deficient thyroid hormones (TH) disrupt different neurochemical, morphological and functional aspects in the cerebral cortex of 15-day-old offspring. Our results showing decreased glutamine synthetase (GS) activity and Ca2+ overload in the cerebral cortex of hypothyroid pups suggest misregulated glutamate metabolism associated with developmentally induced TH deficiency. The 14C-MeAIB accumulation indicates upregulated System A activity and glutamine uptake by neurons. Energy metabolism in hypothyroid cortical slices was preserved, as demonstrated by unaltered glucose metabolism. We also found upregulated acetylcholinesterase activity, depleting acetylcholine from the synaptic cleft, pointing to disrupted cholinergic system. Increased reactive oxygen species (ROS) generation, lipid peroxidation, glutathione (GSH) depletion, which were associated with glutathione peroxidase, superoxide dismutase and gamma-glutamyltransferase downregulation suggest redox imbalance. Disrupted astrocyte cytoskeleton was evidenced by downregulated and hyperphosphorylated glial fibrillary acidic protein (GFAP). Morphological and structural characterization of the sensorimotor cerebral cortex (SCC) showed unaltered thickness of the SCC. However, decreased size of neurons on the layers II & III and IV in the right SCC and increased NeuN positive neurons in specific SCC layers, suggest that they are differently affected by the low TH levels during neurodevelopment. Hypothyroid pups presented increased number of foot-faults in the gridwalk test indicating affected motor functions. Taken together, our results show that congenital hypothyroidism disrupts glutamatergic and cholinergic neurotransmission, Ca2+ equilibrium, redox balance, cytoskeleton integrity, morphological and functional aspects in the cerebral cortex of young rats.


Hypothyroidism/chemically induced , Sensorimotor Cortex/enzymology , Acetylcholinesterase/metabolism , Animals , Animals, Newborn , Antigens, Nuclear/metabolism , Behavior, Animal , Biological Transport , Body Composition , Cells, Cultured , Cerebral Cortex/enzymology , Female , Glial Fibrillary Acidic Protein/metabolism , Glucose/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamic Acid/metabolism , Hypothyroidism/blood , Hypothyroidism/physiopathology , L-Lactate Dehydrogenase/metabolism , Molecular Docking Simulation , Motor Activity , Nerve Tissue Proteins/metabolism , Oxidation-Reduction , Phosphorylation , Propylthiouracil , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/metabolism , Thyroid Gland/metabolism , Thyroid Hormones/blood
3.
Metab Brain Dis ; 33(3): 693-704, 2018 06.
Article En | MEDLINE | ID: mdl-29288365

Although methylphenidate (MPH) is ubiquitously prescribed to children and adolescents, the consequences of chronic utilization of this psychostimulant are poorly understood. In this study, we investigated the effects of MPH on cytoskeletal homeostasis and lipid content in rat hippocampus. Wistar rats received intraperitoneal injections of MPH (2.0 mg/kg) or saline solution (controls), once a day, from the 15th to the 44th day of age. Results showed that MPH provoked hypophosphorylation of glial fibrillary acidic protein (GFAP) and reduced its immunocontent. Middle and high molecular weight neurofilament subunits (NF-M, NF-H) were hypophosphorylated by MPH on KSP repeat tail domains, while NFL, NFM and NFH immunocontents were not altered. MPH increased protein phosphatase 1 (PP1) and 2A (PP2A) immunocontents. MPH also decreased the total content of ganglioside and phospholipid, as well as the main brain gangliosides (GM1, GD1a, and GD1b) and the major brain phospholipids (sphingomyelin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, and phosphatidylserine). Total cholesterol content was also reduced in the hippocampi of juvenile rats treated with MPH. These results provide evidence that disruptions of cytoskeletal and lipid homeostasis in hippocampus of juvenile rats are triggers by chronic MPH treatment and present a new basis for understanding the effects and consequences associated with chronic use of this psychostimulant during the development of the central nervous system.


Cytoskeleton/drug effects , Hippocampus/drug effects , Homeostasis/drug effects , Lipid Metabolism/drug effects , Methylphenidate/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism , Lipids , Male , Rats, Wistar
4.
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
5.
Mol Neurobiol ; 55(6): 5111-5124, 2018 Jun.
Article En | MEDLINE | ID: mdl-28840509

Kynurenic acid (KYNA) is a neuroactive metabolite of tryptophan known to modulate a number of mechanisms involved in neural dysfunction. Although its activity in the brain has been widely studied, the effect of KYNA counteracting the actions of quinolinic acid (QUIN) remains unknown. The present study aims at describing the ability of 100 µM KYNA preventing cytoskeletal disruption provoked by QUIN in astrocyte/neuron/microglia mixed culture. KYNA totally preserved cytoskeletal organization, cell morphology, and redox imbalance in mixed cultures exposed to QUIN. However, KYNA partially prevented morphological alteration in isolated primary astrocytes and failed to protect the morphological alterations of neurons caused by QUIN exposure. Moreover, KYNA prevented QUIN-induced microglial activation and upregulation of ionized calcium-binding adapter molecule 1 (Iba-1) and partially preserved tumor necrosis factor-α (TNF-α) level in mixed cultures. TNF-α level was also partially preserved in astrocytes. In addition to the mechanisms dependent on redox imbalance and microglial activation, KYNA prevented downregulation of connexin-43 and the loss of functionality of gap junctions (GJs), preserving cell-cell contact, cytoskeletal organization, and cell morphology in QUIN-treated cells. Furthermore, the toxicity of QUIN targeting the cytoskeleton of mixed cultures was not prevented by the N-methyl-D-aspartate (NMDA) antagonist MK-801. We suggest that KYNA protects the integrity of the cytoskeleton of mixed cultures by complex mechanisms including modulating microglial activation preventing oxidative imbalance and misregulated GJs leading to disrupted cytoskeleton in QUIN-treated cells. This study contributed to elucidate the molecular basis of KYNA protection against QUIN toxicity.


Corpus Striatum/pathology , Cytoskeleton/metabolism , Kynurenic Acid/pharmacology , Quinolinic Acid/toxicity , Animals , Antioxidants/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Communication/drug effects , Cells, Cultured , Cytoskeleton/drug effects , Dizocilpine Maleate/pharmacology , Gap Junctions/drug effects , Gap Junctions/metabolism , Microglia/drug effects , Microglia/metabolism , Models, Biological , N-Methylaspartate/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Rats, Wistar , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
6.
Mol Neurobiol ; 55(5): 4362-4372, 2018 May.
Article En | MEDLINE | ID: mdl-28647871

Cytoskeletal proteins are increasingly recognized as having important roles as a target of the action of different neurotoxins. In the last years, several works of our group have shown that quinolinic acid (QUIN) was able to disrupt the homeostasis of the cytoskeleton of neural cells and this was associated with cell dysfunction and neurodegeneration. QUIN is an excitotoxic metabolite of tryptophan metabolism and its accumulation is associated with several neurodegenerative diseases. In the present review, we provide a comprehensive view of the actions of QUIN upstream of glutamate receptors, eliciting kinase/phosphatase signaling cascades that disrupt the homeostasis of the phosphorylation system associated with intermediate filament proteins of astrocytes and neurons. We emphasize the critical role of calcium in these actions and the evidence that misregulated cytoskeleton takes part of the cell response to the injury resulting in neurodegeneration in different brain regions, disrupted cell signaling in acute tissue slices, and disorganized cytoskeleton with altered cell morphology in primary cultures. We also discuss the interplay among misregulated cytoskeleton, oxidative stress, and cell-cell contact through gap junctions mediating the quinolinic acid injury in rat brain. The increasing amount of cross talks identified between cytoskeletal proteins and cellular signaling cascades reinforces the exciting possibility that cytoskeleton could be a new target in the neurotoxicity of QUIN and further studies will be necessary to develop strategies to protect the cytoskeleton and counteracts the cytotoxicity of this metabolite.


Cytoskeleton/metabolism , Neurotoxins/toxicity , Quinolinic Acid/toxicity , Animals , Cytoskeleton/drug effects , Disease Models, Animal , Intermediate Filaments/drug effects , Intermediate Filaments/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology
7.
Arch Toxicol ; 92(1): 513-527, 2018 Jan.
Article En | MEDLINE | ID: mdl-28821999

In this study, we assessed some hippocampal signaling cascades and behavioral impairments in 30-day-old rat pups prenatally exposed to methylmercury (MeHg). Pregnant rats were exposed to 1.0 or 2.0 mg/kg MeHg by gavage in alternated days from gestational day 5 until parturition. We found increased anxiety-like and decreased exploration behavior evaluated by open field test and deficit of both short- and long-term memories by novel object recognition task, respectively, in MeHg-treated pups. Downregulated PI3K/Akt/mTOR pathway and activated/hypophosphorylated (Ser9) GSK3ß in MeHg-treated pups could be upstream of hyperphosphorylated Tau (Ser396) destabilizing microtubules and contributing to neural dysfunction in the hippocampus of these rats. Hyperphosphorylated/activated p38MAPK and downregulated phosphoErk1/2 support a role for mitogen-activated protein kinase (MAPK) cascade on MeHg neurotoxicity. Decreased receptor of advanced glycation end products (RAGE) immunocontent supports the assumption that downregulated RAGE/Erk1/2 pathway could be involved in hypophosphorylated lysine/serine/proline (KSP) repeats on neurofilament subunits and disturbed axonal transport. Downregulated myelin basic protein (MBP), the major myelin protein, is compatible with dysmyelination and neurofilament hypophosphorylation. Increased glial fibrillary acidic protein (GFAP) levels suggest reactive astrocytes, and active apoptotic pathways BAD/BCL-2, BAX/BCL-XL, and caspase 3 suggest cell death. Taken together, our findings get light on important signaling mechanisms that could underlie the behavioral deficits in 30-day-old pups prenatally exposed to MeHg.


Hippocampus/growth & development , Methylmercury Compounds/toxicity , Neurotoxicity Syndromes/etiology , Prenatal Exposure Delayed Effects , Signal Transduction/drug effects , Animals , Behavior, Animal/drug effects , Female , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Lipid Peroxidation/drug effects , MAP Kinase Signaling System/drug effects , Male , Memory/drug effects , Neurotoxicity Syndromes/pathology , Phosphatidylinositol 3-Kinases/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/metabolism , Rats, Wistar
8.
Neuromolecular Med ; 19(2-3): 345-356, 2017 Sep.
Article En | MEDLINE | ID: mdl-28689355

The objective of study was to investigate changes caused by ovariectomy (OVX) on aversive and non-aversive memories, as well as on cytoskeleton phosphorylating system and on vitamin D receptor (VDR) immunocontent in hippocampus. The neuroprotective role of vitamin D was also investigated. Ninety-day-old female Wistar rats were divided into four groups: SHAM, OVX, VITAMIN D and OVX + VITAMIN D; 30 days after the OVX, vitamin D supplementation (500 IU/kg), by gavage, for 30 days was started. Results showed that OVX impaired short-term and long-term recognition, and long-term aversive memories. OVX altered hippocampal cytoskeleton phosphorylating system, evidenced by the hyperphosphorylation of glial fibrillary acidic protein (GFAP), low molecular weight neurofilament subunit (NFL), medium molecular weight neurofilament subunit (NFM) and high molecular weight neurofilament subunit (NFH), and increased the immunocontent of c-Jun N-terminal protein kinases (JNK), Ca2+/calmodulin-dependent protein kinase II (PKCaMII) and of the sites phosphorylated lysine-serine-proline (KSP) repeats, Ser55 and Ser57. Vitamin D reversed the effects caused by OVX on cytoskeleton in hippocampus, but it was not able to reverse the effects on memory.


Cholecalciferol/therapeutic use , Cytoskeleton/drug effects , Hippocampus/drug effects , Memory Disorders/drug therapy , Neuroprotective Agents/therapeutic use , Ovariectomy/adverse effects , Animals , Avoidance Learning/drug effects , Cholecalciferol/pharmacology , Cytoskeletal Proteins/metabolism , Drug Evaluation, Preclinical , Exploratory Behavior/drug effects , Female , Hippocampus/metabolism , Hippocampus/pathology , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Phosphorylation , Protein Processing, Post-Translational/drug effects , Random Allocation , Rats , Rats, Wistar
9.
Toxicology ; 379: 1-11, 2017 03 15.
Article En | MEDLINE | ID: mdl-28137618

Diphenylditelluride (PhTe)2 is a neurotoxin that disrupts cytoskeletal homeostasis. We are showing that different concentrations of (PhTe)2 caused hypophosphorylation of glial fibrillary acidic protein (GFAP), vimentin and neurofilament subunits (NFL, NFM and NFH) and altered actin organization in co-cultured astrocytes and neurons from cerebral cortex of rats. These mechanisms were mediated by N-methyl-d-aspartate (NMDA) receptors without participation of either L-type voltage-dependent calcium channels (L-VDCC) or metabotropic glutamate receptors. Upregulated Ca2+ influx downstream of NMDA receptors activated Ca2+-dependent protein phosphatase 2B (PP2B) causing hypophosphorylation of astrocyte and neuron IFs. Immunocytochemistry showed that hypophosphorylated intermediate filaments (IF) failed to disrupt their organization into the cytoskeleton. However, phalloidin-actin-FITC stained cytoskeleton evidenced misregulation of actin distribution, cell spreading and increased stress fibers in astrocytes. ßIII tubulin staining showed that neurite meshworks are not altered by (PhTe)2, suggesting greater susceptibility of astrocytes than neurons to (PheTe)2 toxicity. These findings indicate that signals leading to IF hypophosphorylation fail to disrupt the cytoskeletal IF meshwork of interacting astrocytes and neurons in vitro however astrocyte actin network seems more susceptible. Our findings support that intracellular Ca2+ is one of the crucial signals that modulate the action of (PhTe)2 in co-cultured astrocytes and neurons and highlights the cytoskeleton as an end-point of the neurotoxicity of this compound. Cytoskeletal misregulation is associated with cell dysfunction, therefore, the understanding of the molecular mechanisms mediating the neurotoxicity of this compound is a matter of increasing interest since tellurium compounds are increasingly released in the environment.


Astrocytes/drug effects , Benzene Derivatives/toxicity , Cell Communication , Cytoskeleton/drug effects , Neurons/drug effects , Organometallic Compounds/toxicity , Animals , Astrocytes/physiology , Calcium/metabolism , Coculture Techniques , Neurons/physiology , Phosphorylation , Rats
10.
Mol Neurobiol ; 54(8): 5752-5767, 2017 10.
Article En | MEDLINE | ID: mdl-27660266

In the present work, we focused on mechanisms of methylmercury (MeHg) toxicity in primary astrocytes and neurons of rats. Cortical astrocytes and neurons exposed to 0.5-5 µM MeHg present a link among morphological alterations, glutathione (GSH) depletion, glutamate dyshomeostasis, and cell death. Disrupted neuronal cytoskeleton was assessed by decreased neurite length and neurite/neuron ratio. Astrocytes presented reorganization of actin and glial fibrillary acidic protein (GFAP) networks and reduced cytoplasmic area. Glutamate uptake and Na+K+ATPase activity in MeHg-treated astrocytes were preserved; however, downregulated EAAC1-mediated glutamate uptake was associated with impaired Na+K+ATPase activity in neurons. Oxidative imbalance was found in astrocytes and neurons through increased 2'7'-dichlorofluorescein (DCF) production and misregulated superoxide dismutase (SOD), catalase (CAT), and glutathione reductase (GPX) activities. Glutathione (GSH) levels were downregulated in both astrocytes and neurons. MeHg reduced neuronal viability and induced caspase 3-dependent apoptosis together with downregulated PI3K/Akt pathway. In astrocytes, necrotic death was associated with increased TNF-α and JNK/MAPK activities. Cytoskeletal remodeling and cell death were fully prevented in astrocytes and neurons by GSH, but not melatonin or Trolox supplementation. These findings support a role for depleted GSH in the cytotoxicity of MeHg leading to disruption of the cytoskeleton and cell death. Moreover, in neurons, glutamate antagonists also prevented cytoskeletal disruption and neuronal death. We propose that cytoskeleton is an end point in MeHg cytotoxicity. Oxidative imbalance and glutamate mechanisms mediate MeHg cytoskeletal disruption and apoptosis in neurons. Otherwise, redox imbalance and glutamate-independent mechanisms disrupted the cytoskeleton and induced necrosis in MeHg-exposed astrocyte.


Astrocytes/drug effects , Cytoskeleton/drug effects , Methylmercury Compounds/pharmacology , Neurons/metabolism , Animals , Animals, Newborn , Astrocytes/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Female , Neurons/drug effects , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Rats, Wistar , Reactive Oxygen Species/metabolism
11.
Biochim Biophys Acta ; 1863(12): 3001-3014, 2016 12.
Article En | MEDLINE | ID: mdl-27663072

QUIN is a glutamate agonist playing a role in the misregulation of the cytoskeleton, which is associated with neurodegeneration in rats. In this study, we focused on microglial activation, FGF2/Erk signaling, gap junctions (GJs), inflammatory parameters and redox imbalance acting on cytoskeletal dynamics of the in QUIN-treated neural cells of rat striatum. FGF-2/Erk signaling was not altered in QUIN-treated primary astrocytes or neurons, however cytoskeleton was disrupted. In co-cultured astrocytes and neurons, QUIN-activated FGF2/Erk signaling prevented the cytoskeleton from remodeling. In mixed cultures (astrocyte, neuron, microglia), QUIN-induced FGF-2 increased level failed to activate Erk and promoted cytoskeletal destabilization. The effects of QUIN in mixed cultures involved redox imbalance upstream of Erk activation. Decreased connexin 43 (Cx43) immunocontent and functional GJs, was also coincident with disruption of the cytoskeleton in primary astrocytes and mixed cultures. We postulate that in interacting astrocytes and neurons the cytoskeleton is preserved against the insult of QUIN by activation of FGF-2/Erk signaling and proper cell-cell interaction through GJs. In mixed cultures, the FGF-2/Erk signaling is blocked by the redox imbalance associated with microglial activation and disturbed cell communication, disrupting the cytoskeleton. Thus, QUIN signal activates differential mechanisms that could stabilize or destabilize the cytoskeleton of striatal astrocytes and neurons in culture, and glial cells play a pivotal role in these responses preserving or disrupting a combination of signaling pathways and cell-cell interactions. Taken together, our findings shed light into the complex role of the active interaction of astrocytes, neurons and microglia in the neurotoxicity of QUIN.


Astrocytes/drug effects , Cytoskeleton/drug effects , Excitatory Amino Acid Agonists/toxicity , Microglia/drug effects , Quinolinic Acid/toxicity , Animals , Astrocytes/cytology , Astrocytes/metabolism , Cell Communication/drug effects , Coculture Techniques , Connexin 43/genetics , Connexin 43/metabolism , Corpus Striatum/cytology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gap Junctions/drug effects , Gap Junctions/metabolism , Gap Junctions/ultrastructure , Gene Expression Regulation , MAP Kinase Signaling System , Microglia/cytology , Microglia/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oxidation-Reduction , Pregnancy , Primary Cell Culture , Rats , Rats, Wistar
12.
Biochim Biophys Acta ; 1860(11 Pt A): 2510-2520, 2016 11.
Article En | MEDLINE | ID: mdl-27475002

BACKGROUND: Diphenylditelluride (PhTe)2 is a potent neurotoxin disrupting the homeostasis of the cytoskeleton. METHODS: Cultured astrocytes and neurons were incubated with (PhTe)2, receptor antagonists and enzyme inhibitors followed by measurement of the incorporation of [32P]orthophosphate into intermediate filaments (IFs). RESULTS: (PhTe)2 caused hyperphosphorylation of glial fibrillary acidic protein (GFAP), vimentin and neurofilament subunits (NFL, NFM and NFH) from primary astrocytes and neurons, respectively. These mechanisms were mediated by N-methyl-d-aspartate (NMDA) receptors, L-type voltage-dependent calcium channels (L-VDCCs) as well as metabotropic glutamate receptors upstream of phospholipase C (PLC). Upregulated Ca(2+) influx activated protein kinase A (PKA) and protein kinase C (PKC) in astrocytes causing hyperphosphorylation of GFAP and vimentin. Hyperphosphorylated (IF) together with RhoA-activated stress fiber formation, disrupted the cytoskeleton leading to altered cell morphology. In neurons, the high intracellular Ca(2+) levels activated the MAPKs, Erk and p38MAPK, beyond PKA and PKC, provoking hyperphosphorylation of NFM, NFH and NFL. CONCLUSIONS: Our findings support that intracellular Ca(2+) is one of the crucial signals that modulate the action of (PhTe)2 in isolated cortical astrocytes and neurons modulating the response of the cytoskeleton against the insult. GENERAL SIGNIFICANCE: Cytoskeletal misregulation is associated with neurodegeneration. This compound could be a valuable tool to induce molecular changes similar to those found in different pathologies of the brain.


Actin Cytoskeleton/metabolism , Astrocytes/drug effects , Benzene Derivatives/pharmacology , Calcium Signaling , Neurons/drug effects , Organometallic Compounds/pharmacology , Animals , Astrocytes/metabolism , Benzene Derivatives/toxicity , Cells, Cultured , Neurons/metabolism , Organometallic Compounds/toxicity , Rats , Rats, Wistar
13.
Neurotox Res ; 30(2): 138-49, 2016 08.
Article En | MEDLINE | ID: mdl-26936604

In the present work, we studied the effects of toxic ammonia levels on the cytoskeleton of neural cells, with emphasis in the homeostasis of the phosphorylating system associated with the intermediate filaments (IFs). We used in vivo and in vitro models of acute hyperammonemia in 10- and 21-day-old rats. In the in vivo model, animals were intraperitoneally injected with ammonium acetate (7 mmol/Kg), and the phosphorylation level of the cytoskeletal proteins was analyzed in the cerebral cortex and hippocampus 30 and 60 min after injection. The injected ammonia altered the IF phosphorylation of astrocytes (GFAP and vimentin) and neurons (neurofilament subunits of low, middle, and high molecular weight, respectively: NFL, NFM, and NFH) from cerebral cortex of 21-day-old rats. This was a transitory effect observed 30 min after injection, recovering 30 min afterward. Phosphorylation was not altered in the cerebral cortex of 10-day-old pups. The homeostasis of hippocampal IFs was preserved at the studied ages and times. In the in vitro model, cortical slices of 10- and 21-day-old rats were incubated with 0.5, 1, or 5 mM NH4Cl, and the phosphorylation level of the IF proteins was analyzed after 30 min. The IF phosphorylation was not altered in cortical slices of 10-day-old rats; however, in cortical slices of 21-day-old pups, 5 mM NH4Cl induced hypophosphorylation of GFAP and vimentin, preserving neurofilament phosphorylation levels. Hypophosphorylation was mediated by the protein phosphatases 1 (PP1) and 2B (PP2B), and this event was associated with Ca(2+) influx via N-methyl-D-aspartate (NMDA) glutamate receptors. The aim of this study is to show that acute ammonia toxicity targets the phosphorylating system of IFs in the cerebral cortex of rats in a developmentally regulated manner, and NMDA-mediated Ca(2+) signaling plays a central role in this mechanism. We propose that the disruption of cytoskeletal homeostasis could be an endpoint of the acute hyperammonemia in the developing brain. We believe that these results contribute for better understanding the molecular basis of the ammonia toxicity in brain.


Calcineurin/metabolism , Cerebral Cortex/metabolism , Hyperammonemia/metabolism , Intermediate Filaments/metabolism , N-Methylaspartate/metabolism , Protein Phosphatase 1/metabolism , Acetates , Aging/metabolism , Ammonium Chloride , Animals , Astrocytes/metabolism , Calcium/metabolism , Calcium Signaling/physiology , Disease Models, Animal , Hippocampus/metabolism , Neurons/metabolism , Phosphorylation/physiology , Rats, Wistar , Time Factors , Tissue Culture Techniques
14.
Mol Neurobiol ; 53(2): 1065-1079, 2016 Mar.
Article En | MEDLINE | ID: mdl-25579384

Hyperprolinemias are inherited disorder of proline (Pro) metabolism. Patients affected may present neurological manifestations, but the mechanisms of neural excitotoxicity elicited by hyperprolinemia are far from being understood. Considering that the astrocytes are important players in neurological disorders, the aim of the present work was to study the effects 1 mM Pro on glutamatergic and inflammatory parameters in cultured astrocytes from cerebral cortex of rats, exploring some molecular mechanisms underlying the disrupted homeostasis of astrocytes exposed to this toxic Pro concentration. We showed that cortical astrocytes of rats exposed to 1 mM Pro presented significantly elevated extracellular glutamate and glutamine levels, suggesting glutamate excitotoxicity. The excess of glutamate elicited by Pro together with increased glutamate uptake and upregulated glutamine synthetase (GS) activity supported misregulated glutamate homeostasis in astrocytic cells. High Pro levels also induced production/release of pro-inflammatory cytokines TNF-α, IL-1ß, and IL-6. We also evidenced misregulation of cholinergic anti-inflammatory system with increased acetylcholinesterase (AChE) activity and decreased acetylcholine (ACh) levels, contributing to the inflammatory status in Pro-treated astrocytes. Our findings highlighted a crosstalk among disrupted glutamate homeostasis, cholinergic mechanisms, and inflammatory cytokines, since ionotropic (DL-AP5 and CNQX) and metabotropic (MCPG and MPEP) glutamate antagonists were able to restore the extracellular glutamate and glutamine levels; downregulate TNFα and IL6 production/release, modulate GS and AChE activities; and restore ACh levels. Otherwise, the non-steroidal anti-inflammatory drugs nimesulide, acetylsalicylic acid, ibuprofen, and diclofenac sodium decreased the extracellular glutamate and glutamine levels, downregulated GS and AChE activities, and restored ACh levels in Pro-treated astrocytes. Altogether, our results evidence that the vulnerability of metabolic homeostasis in cortical astrocytes might have important implications in the neurotoxicity of Pro.


Astrocytes/metabolism , Choline/metabolism , Glutamic Acid/metabolism , Homeostasis/drug effects , Inflammation/metabolism , Inflammation/pathology , Proline/pharmacology , Acetylcholinesterase/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Astrocytes/drug effects , Cerebral Cortex/cytology , Cytokines/metabolism , Glutamate-Ammonia Ligase/metabolism , Glutamine/metabolism , Rats, Wistar
15.
Alcohol ; 49(7): 665-74, 2015 Nov.
Article En | MEDLINE | ID: mdl-26314629

Ethanol exposure to offspring during pregnancy and lactation leads to developmental disorders, including central nervous system dysfunction. In the present work, we have studied the effect of chronic ethanol exposure during pregnancy and lactation on the phosphorylating system associated with the astrocytic and neuronal intermediate filament (IF) proteins: glial fibrillary acidic protein (GFAP), and neurofilament (NF) subunits of low, medium, and high molecular weight (NFL, NFM, and NFH, respectively) in 9- and 21-day-old pups. Female rats were fed with 20% ethanol in their drinking water during pregnancy and lactation. The homeostasis of the IF phosphorylation was not altered in the cerebral cortex, cerebellum, or hippocampus of 9-day-old pups. However, GFAP, NFL, and NFM were hyperphosphorylated in the hippocampus of 21-day-old pups. PKA had been activated in the hippocampus, and Ser55 in the N-terminal region of NFL was hyperphosphorylated. In addition, JNK/MAPK was activated and KSP repeats in the C-terminal region of NFM were hyperphosphorylated in the hippocampus of 21-day-old pups. Decreased NFH immunocontent but an unaltered total NFH/phosphoNFH ratio suggested altered stoichiometry of NFs in the hippocampus of ethanol-exposed 21-day-old pups. In contrast to the high susceptibility of hippocampal cytoskeleton in developing rats, the homeostasis of the cytoskeleton of ethanol-fed adult females was not altered. Disruption of the cytoskeletal homeostasis in neural cells supports the view that regions of the brain are differentially vulnerable to alcohol insult during pregnancy and lactation, suggesting that modulation of JNK/MAPK and PKA signaling cascades target the hippocampal cytoskeleton in a window of vulnerability in 21-day-old pups. Our findings are relevant, since disruption of the cytoskeleton in immature hippocampus could contribute to later hippocampal damage associated with ethanol toxicity.


Central Nervous System Depressants/toxicity , Cytoskeleton/drug effects , Ethanol/toxicity , Hippocampus/drug effects , Lactation , Animals , Animals, Newborn , Body Weight/drug effects , Cyclic AMP-Dependent Protein Kinases/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Energy Intake/drug effects , Female , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/embryology , Homeostasis , Intermediate Filaments/drug effects , MAP Kinase Signaling System/drug effects , Neurofilament Proteins/metabolism , Phosphorylation , Pregnancy , Rats , Rats, Wistar
16.
Neurotox Res ; 28(3): 239-52, 2015 Oct.
Article En | MEDLINE | ID: mdl-26174040

Neurological symptoms and cerebral abnormalities are commonly observed in patients with 3-hydroxy-3-methylglutaryl-CoA lyase (HMG lyase) deficiency, which is biochemically characterized by predominant tissue accumulation of 3-hydroxy-3-methylglutaric (HMG), 3-methylglutaric (MGA), and 3-methylglutaconic (MGT) acids. Since the pathogenesis of this disease is poorly known, the present study evaluated the effects of these compounds on the cytoskeleton phosphorylating system in rat brain. HMG, MGA, and MGT caused hypophosphorylation of glial fibrillary acidic protein (GFAP) and of the neurofilament subunits NFL, NFM, and NFH. HMG-induced hypophosphorylation was mediated by inhibiting the cAMP-dependent protein kinase (PKA) on Ser55 residue of NFL and c-Jun kinase (JNK) by acting on KSP repeats of NFM and NFH subunits. We also evidenced that the subunit NR2B of NMDA receptor and Ca(2+) was involved in HMG-elicited hypophosphorylation of cytoskeletal proteins. Furthermore, the antioxidants L-NAME and TROLOX fully prevented both the hypophosphorylation and the inhibition of PKA and JNK caused by HMG, suggesting that oxidative damage may underlie these effects. These findings indicate that the main metabolites accumulating in HMG lyase deficiency provoke hypophosphorylation of cytoskeleton neural proteins with the involvement of NMDA receptors, Ca(2+), and reactive species. It is presumed that these alterations may contribute to the neuropathology of this disease.


Acetyl-CoA C-Acetyltransferase/deficiency , Amino Acid Metabolism, Inborn Errors/metabolism , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , Cytoskeletal Proteins/metabolism , Oxidative Stress/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Acetyl-CoA C-Acetyltransferase/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Blotting, Western , Calcium/metabolism , Cell Survival/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Corpus Striatum/drug effects , Corpus Striatum/growth & development , Corpus Striatum/pathology , Oxidative Stress/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Rats, Wistar
17.
J Neurosci Res ; 93(2): 268-84, 2015 Feb.
Article En | MEDLINE | ID: mdl-25306914

Quinolinic acid (QUIN) is an endogenous metabolite of the kynurenine pathway involved in several neurological disorders. Among the several mechanisms involved in QUIN-mediated toxicity, disruption of the cytoskeleton has been demonstrated in striatally injected rats and in striatal slices. The present work searched for the actions of QUIN in primary striatal neurons. Neurons exposed to 10 µM QUIN presented hyperphosphorylated neurofilament (NF) subunits (NFL, NFM, and NFH). Hyperphosphorylation was abrogated in the presence of protein kinase A and protein kinase C inhibitors H89 (20 µM) and staurosporine (10 nM), respectively, as well as by specific antagonists to N-methyl-D-aspartate (50 µM DL-AP5) and metabotropic glutamate receptor 1 (100 µM MPEP). Also, intra- and extracellular Ca(2+) chelators (10 µM BAPTA-AM and 1 mM EGTA, respectively) and Ca(2+) influx through L-type voltage-dependent Ca(2+) channel (10 µM verapamil) are implicated in QUIN-mediated effects. Cells immunostained for the neuronal markers ßIII-tubulin and microtubule-associated protein 2 showed altered neurite/neuron ratios and neurite outgrowth. NF hyperphosphorylation and morphological alterations were totally prevented by conditioned medium from QUIN-treated astrocytes. Cocultured astrocytes and neurons interacted with one another reciprocally, protecting them against QUIN injury. Cocultured cells preserved their cytoskeletal organization and cell morphology together with unaltered activity of the phosphorylating system associated with the cytoskeleton. This article describes cytoskeletal disruption as one of the most relevant actions of QUIN toxicity in striatal neurons in culture with soluble factors secreted by astrocytes, with neuron-astrocyte interaction playing a role in neuroprotection.


Astrocytes/physiology , Cell Communication/physiology , Corpus Striatum/cytology , Cytoskeleton/metabolism , Homeostasis/drug effects , Neurons/drug effects , Quinolinic Acid/pharmacology , Animals , Animals, Newborn , Astrocytes/chemistry , Cell Communication/drug effects , Cell Survival/drug effects , Cells, Cultured , Chelating Agents/pharmacology , Coculture Techniques , Culture Media, Conditioned/pharmacology , Dose-Response Relationship, Drug , Egtazic Acid/analogs & derivatives , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Phosphorylation/drug effects , Pregnancy , Rats , Rats, Wistar , Valine/analogs & derivatives , Valine/pharmacology
18.
Brain Res ; 1577: 1-10, 2014 Aug 19.
Article En | MEDLINE | ID: mdl-24976581

Quinolinic acid (QUIN) is a neuroactive metabolite of the kinurenine pathway, considered to be involved in aging and some neurodegenerative disorders, including Huntington׳s disease. In the present work we have studied the long-lasting effect of acute intrastriatal injection of QUIN (150 nmol/0.5 µL) in 30 day-old rats on the phosphorylating system associated with the astrocytic and neuronal intermediate filament (IF) proteins: glial fibrillary acidic protein (GFAP), and neurofilament (NF) subunits (NFL, NFM and NFH) respectively, until 21 days after injection. The acute administration of QUIN altered the homeostasis of IF phosphorylation in a selective manner, progressing from striatum to cerebral cortex and hippocampus. Twenty four hours after QUIN injection, the IFs were hyperphosphorylated in the striatum. This effect progressed to cerebral cortex causing hypophosphorylation at day 14 and appeared in the hippocampus as hyperphosphorylation at day 21 after QUIN infusion. PKA and PKCaMII have been activated in striatum and hippocampus, since Ser55 and Ser57 in NFL head domain were hyperphosphorylated. However, MAPKs (Erk1/2, JNK and p38MAPK) were hyperphosphorylated/activated only in the hippocampus, suggesting different signaling mechanisms in these two brain structures during the first weeks after QUIN infusion. Also, protein phosphatase 1 (PP1) and 2B (PP2B)-mediated hypophosphorylation of the IF proteins in the cerebral cortex 14 after QUIN injection reinforce the selective signaling mechanisms in different brain structures. Increased GFAP immunocontent in the striatum and cerebral cortex 24h and 14 days after QUIN injection respectively, suggests reactive astrocytes in these brain regions. We propose that disruption of cytoskeletal homeostasis in neural cells takes part of the long-lasting molecular mechanisms of QUIN toxicity in adolescent rats, showing selective and progressive misregulation of the signaling mechanisms targeting the IF proteins in the striatum, cerebral cortex and hippocampus with important implications for brain function.


Cerebral Cortex/physiopathology , Corpus Striatum/physiopathology , Cytoskeleton/physiology , Hippocampus/physiopathology , Quinolinic Acid/metabolism , Aging , Animals , Astrocytes/physiology , Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glial Fibrillary Acidic Protein/metabolism , Homeostasis/physiology , Neurofilament Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Rats, Wistar , Time Factors
19.
Oxid Med Cell Longev ; 2014: 458601, 2014.
Article En | MEDLINE | ID: mdl-25050142

Evidence from our group supports that diphenyl ditelluride (PhTe)2 neurotoxicity depends on modulation of signaling pathways initiated at the plasma membrane. The (PhTe)2-evoked signal is transduced downstream of voltage-dependent Ca(2+) channels (VDCC), N-methyl-D-aspartate receptors (NMDA), or metabotropic glutamate receptors activation via different kinase pathways (protein kinase A, phospholipase C/protein kinase C, mitogen-activated protein kinases (MAPKs), and Akt signaling pathway). Among the most relevant cues of misregulated signaling mechanisms evoked by (PhTe)2 is the cytoskeleton of neural cells. The in vivo and in vitro exposure to (PhTe)2 induce hyperphosphorylation/hypophosphorylation of neuronal and glial intermediate filament (IF) proteins (neurofilaments and glial fibrillary acidic protein, resp.) in different brain structures of young rats. Phosphorylation of IFs at specific sites modulates their association/disassociation and interferes with important physiological roles, such as axonal transport. Disrupted cytoskeleton is a crucial marker of neurodegeneration and is associated with reactive astrogliosis and apoptotic cell death. This review focuses the current knowledge and important results on the mechanisms of (PhTe)2 neurotoxicity with special emphasis on the cytoskeletal proteins and their differential regulation by kinases/phosphatases and Ca(2+)-mediated mechanisms in developmental rat brain. We propose that the disrupted cytoskeletal homeostasis could support brain damage provoked by this neurotoxicant.


Benzene Derivatives/toxicity , Cytoskeleton/pathology , Neurotoxins/toxicity , Organometallic Compounds/toxicity , Signal Transduction/drug effects , Animals , Cytoskeleton/drug effects , Humans , Intermediate Filaments/drug effects , Intermediate Filaments/metabolism , Nerve Degeneration/pathology
20.
Exp Cell Res ; 322(2): 313-23, 2014 Apr 01.
Article En | MEDLINE | ID: mdl-24583400

Quinolinic acid (QUIN) is a glutamate agonist which markedly enhances the vulnerability of neural cells to excitotoxicity. QUIN is produced from the amino acid tryptophan through the kynurenine pathway (KP). Dysregulation of this pathway is associated with neurodegenerative conditions. In this study we treated striatal astrocytes in culture with QUIN and assayed the endogenous phosphorylating system associated with glial fibrillary acidic protein (GFAP) and vimentin as well as cytoskeletal remodeling. After 24h incubation with 100 µM QUIN, cells were exposed to (32)P-orthophosphate and/or protein kinase A (PKA), protein kinase dependent of Ca(2+)/calmodulin II (PKCaMII) or protein kinase C (PKC) inhibitors, H89 (20 µM), KN93 (10 µM) and staurosporin (10nM), respectively. Results showed that hyperphosphorylation was abrogated by PKA and PKC inhibitors but not by the PKCaMII inhibitor. The specific antagonists to ionotropic NMDA and non-NMDA (50 µM DL-AP5 and CNQX, respectively) glutamate receptors as well as to metabotropic glutamate receptor (mGLUR; 50 µM MCPG), mGLUR1 (100 µM MPEP) and mGLUR5 (10 µM 4C3HPG) prevented the hyperphosphorylation provoked by QUIN. Also, intra and extracellular Ca(2+) quelators (1mM EGTA; 10 µM BAPTA-AM, respectively) prevented QUIN-mediated effect, while Ca(2+) influx through voltage-dependent Ca(2+) channel type L (L-VDCC) (blocker: 10 µM verapamil) is not implicated in this effect. Morphological analysis showed dramatically altered actin cytoskeleton with concomitant change of morphology to fusiform and/or flattened cells with retracted cytoplasm and disruption of the GFAP meshwork, supporting misregulation of actin cytoskeleton. Both hyperphosphorylation and cytoskeletal remodeling were reversed 24h after QUIN removal. Astrocytes are highly plastic cells and the vulnerability of astrocyte cytoskeleton may have important implications for understanding the neurotoxicity of QUIN in neurodegenerative disorders.


Actin Cytoskeleton/drug effects , Astrocytes/cytology , Corpus Striatum/cytology , Quinolinic Acid/pharmacology , Actin Cytoskeleton/metabolism , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Calcium/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , Glutamates/metabolism , Immunoenzyme Techniques , Phosphorylation/drug effects , Pregnancy , Rats , Rats, Wistar , Vimentin/metabolism
...