Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-35346791

ABSTRACT

Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor that has recently been implicated in several psychiatric conditions related to monoaminergic dysfunction, such as schizophrenia, substance use disorders, and mood disorders. Although attention-deficit/hyperactivity disorder (ADHD) is also related to changes in monoaminergic neurotransmission, studies that assess whether TAAR1 participates in the neurobiology of ADHD are lacking. We hypothesized that TAAR1 plays an important role in ADHD and might represent a potential therapeutic target. Here, we investigate if TAAR1 modulates behavioral phenotypes in Spontaneously Hypertensive Rats (SHR), the most validated animal model of ADHD, and Wistar Kyoto rats (WKY, used as a control strain). Our results showed that TAAR1 is downregulated in ADHD-related brain regions in SHR compared with WKY. While intracerebroventricular (i.c.v.) administration of the selective TAAR1 antagonist EPPTB impaired cognitive performance in SHR, i.c.v. administration of highly selective TAAR1 full agonist RO5256390 decreased motor hyperactivity, novelty-induced locomotion, and induced an anxiolytic-like behavior. Overall, our findings show that changes in TAAR1 levels/activity underlie behavior in SHR, suggesting that TAAR1 plays a role in the neurobiology of ADHD. Although additional confirmatory studies are required, TAAR1 might be a potential pharmacological target for individuals with this disorder.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Receptors, G-Protein-Coupled , Animals , Anxiety/drug therapy , Attention Deficit Disorder with Hyperactivity/psychology , Behavior, Animal , Cognition , Disease Models, Animal , Hyperkinesis , Psychomotor Agitation , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, G-Protein-Coupled/genetics
2.
Biochim Biophys Acta Mol Cell Res ; 1867(8): 118732, 2020 08.
Article in English | MEDLINE | ID: mdl-32360667

ABSTRACT

Nitric oxide is an important neuromodulator in the CNS, and its production within neurons is modulated by NMDA receptors and requires a fine-tuned availability of L-arginine. We have previously shown that globally inhibiting protein synthesis mobilizes intracellular L-arginine "pools" in retinal neurons, which concomitantly enhances neuronal nitric oxide synthase-mediated nitric oxide production. Activation of NMDA receptors also induces local inhibition of protein synthesis and L-arginine intracellular accumulation through calcium influx and stimulation of eucariotic elongation factor type 2 kinase. We hypothesized that protein synthesis inhibition might also increase intracellular L-arginine availability to induce nitric oxide-dependent activation of downstream signaling pathways. Here we show that nitric oxide produced by inhibiting protein synthesis (using cycloheximide or anisomycin) is readily coupled to AKT activation in a soluble guanylyl cyclase and cGKII-dependent manner. Knockdown of cGKII prevents cycloheximide or anisomycin-induced AKT activation and its nuclear accumulation. Moreover, in retinas from cGKII knockout mice, cycloheximide was unable to enhance AKT phosphorylation. Indeed, cycloheximide also produces an increase of ERK phosphorylation which is abrogated by a nitric oxide synthase inhibitor. In summary, we show that inhibition of protein synthesis is a previously unanticipated driving force for nitric oxide generation and activation of downstream signaling pathways including AKT and ERK in cultured retinal cells. These results may be important for the regulation of synaptic signaling and neuronal development by NMDA receptors as well as for solving conflicting data observed when using protein synthesis inhibitors for studying neuronal survival during development as well in behavior and memory studies.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Nitric Oxide/metabolism , Protein Synthesis Inhibitors/pharmacology , Retina/metabolism , Signal Transduction/drug effects , Animals , Arginine/metabolism , Cell Survival/drug effects , Cells, Cultured , Chick Embryo , Chickens , Cyclic GMP-Dependent Protein Kinase Type II/genetics , Elongation Factor 2 Kinase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Nitrates/metabolism , Nitric Oxide Synthase Type I/metabolism , Nitrites , Phosphorylation
3.
Front Neurosci ; 13: 453, 2019.
Article in English | MEDLINE | ID: mdl-31143097

ABSTRACT

Ascorbate, the reduced form of Vitamin C, is one of the most abundant and important low-molecular weight antioxidants in living tissues. Most animals synthesize vitamin C, but some primates, including humans, have lost this capacity due to disruption in L-gulono-gamma-lactone oxidase gene. Because of this incapacity, those animals must obtain Vitamin C from the diet. Ascorbate is highly concentrated in the central nervous system (CNS), including the retina, and plays essential roles in neuronal physiology. Ascorbate transport into cells is controlled by Sodium Vitamin C Co-Transporters (SVCTs). There are four SVCT isoforms and SVCT2 is the major isoform controlling ascorbate transport in the CNS. Regarding ascorbate release from retinal neurons, Glutamate, by activating its ionotropic receptors leads to ascorbate release via the reversion of SVCT2. Moreover, dopamine, via activation of D1 receptor/cyclic AMP/EPAC2 pathway, also induces ascorbate release via SVCT2 reversion. Because the dopaminergic and glutamatergic systems are interconnected in the CNS, we hypothesized that dopamine could regulate ascorbate release indirectly, via the glutamatergic system. Here we reveal that dopamine increases the release of D-Aspartate from retinal neurons in a way independent on calcium ions and dependent on excitatory amino acid transporters. In addition, dopamine-dependent SVCT2 reversion leading to ascorbate release occurs by activation of AMPA/Kainate receptors and downstream ERK/AKT pathways. Overall, our data reveal a dopamine-to-glutamate signaling that regulates the bioavailability of ascorbate in neuronal cells.

4.
Biochem Pharmacol ; 155: 393-402, 2018 09.
Article in English | MEDLINE | ID: mdl-30031809

ABSTRACT

Chlorogenic acids (CGAs) are a group of phenolic compounds found in worldwide consumed beverages such as coffee and green tea. They are synthesized from an esterification reaction between cinnamic acids, including caffeic (CFA), ferulic and p-coumaric acids with quinic acid (QA), forming several mono- and di-esterified isomers. The most prevalent and studied compounds are 3-O-caffeoylquinic acid (3-CQA), 4-O-caffeoylquinic acid (4-CQA) and 5-O-caffeoylquinic acid (5-CQA), widely described as having antioxidant and cell protection effects. CGAs can also modulate glutamate release from microglia by a mechanism involving a decrease of reactive oxygen species (ROS). Increased energy metabolism is highly associated with enhancement of ROS production and cellular damage. Glutamate can also be used as an energy source by glutamate dehydrogenase (GDH) enzyme, providing α-ketoglutarate to the tricarboxylic acid (TCA) cycle for ATP synthesis. High GDH activity is associated with some disorders, such as schizophrenia and hyperinsulinemia/hyperammonemia syndrome. In line with this, our objective was to investigate the effect of CGAs on GDH activity. We show that CGAs and CFA inhibits GDH activity in dose-dependent manner, reaching complete inhibition at high concentration with IC50 of 52 µM for 3-CQA and 158.2 µM for CFA. Using live imaging confocal microscopy and microplate reader, we observed that 3-CQA and CFA can be transported into neuronal cells by an Na+-dependent mechanism. Moreover, neuronal cells treated with CGAs presented lower intracellular ATP levels. Overall, these data suggest that CGAs have therapeutic potential for treatment of disorders associated with high GDH activity.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Chlorogenic Acid/pharmacology , Glutamate Dehydrogenase/antagonists & inhibitors , Intracellular Fluid/drug effects , Retina/drug effects , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Chick Embryo , Dose-Response Relationship, Drug , Glutamate Dehydrogenase/metabolism , Intracellular Fluid/metabolism , Retina/cytology , Retina/metabolism
5.
Mol Neurobiol ; 55(8): 6816-6833, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29349577

ABSTRACT

Glioblastoma (GBM) is the most common and aggressive malignant primary brain tumor and still lacks effective therapeutic strategies. It has already been shown that old drugs like sulfasalazine (SAS) and valproic acid (VPA) present antitumoral activities in glioma cell lines. SAS has also been associated with a decrease of intracellular glutathione (GSH) levels through a potent inhibition of xc- glutamate/cystine exchanger leading to an antioxidant deprotection. In the same way, VPA was recently identified as a histone deacetylase (HDAT) inhibitor capable of activating tumor suppression genes. As both drugs are widely used in clinical practice and their profile of adverse effects is well known, the aim of our study was to investigate the effects of the combined treatment with SAS and VPA in GBM cell lines. We observed that both drugs were able to reduce cell viability in a dose-dependent manner and the combined treatment potentiated these effects. Combined treatment also increased cell death and inhibited proliferation of GBM cells, while having no effect on human and rat cultured astrocytes. Also, we observed high protein expression of the catalytic subunit of xc- in all the examined GBM cell lines, and treatment with SAS blocked its activity and decreased intracellular GSH levels. Noteworthy, SAS but not VPA was also able to reduce the [14C]-ascorbate uptake. Together, these data indicate that SAS and VPA exhibit a substantial effect on GBM cell's death related to an intracellular oxidative response imbalance, making this combination of drugs a promising therapeutic strategy.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Intracellular Space/metabolism , Sulfasalazine/pharmacology , Valproic Acid/pharmacology , Amino Acid Transport System y+/metabolism , Animals , Ascorbic Acid/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Therapy, Combination , Glutathione/metabolism , Humans , Mesoderm/drug effects , Mesoderm/pathology , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Oxidation-Reduction , Rats , Time Factors
6.
J Neurochem ; 144(4): 408-420, 2018 02.
Article in English | MEDLINE | ID: mdl-29164598

ABSTRACT

Vitamin C (in the reduced form ascorbate or in the oxidized form dehydroascorbate) is implicated in signaling events throughout the central nervous system (CNS). In the retina, a high-affinity transport system for ascorbate has been described and glutamatergic signaling has been reported to control ascorbate release. Here, we investigated the modulatory role played by vitamin C upon glutamate uptake and N-methyl-d-aspartate (NMDA) receptor activation in cultured retinal cells or in intact retinal tissue using biochemical and imaging techniques. We show that both forms of vitamin C, ascorbate or dehydroascorbate, promote an accumulation of extracellular glutamate by a mechanism involving the inhibition of glutamate uptake. This inhibition correlates with the finding that ascorbate promotes a decrease in cell surface levels of the neuronal glutamate transporter excitatory amino acid transporter 3 in retinal neuronal cultures. Interestingly, vitamin C is prone to increase the activity of NMDA receptors but also promotes a decrease in glutamate-stimulated [3 H] MK801 binding and decreases cell membrane content of NMDA receptor glutamate ionotropic receptor subunit 1 (GluN1) subunits. Both compounds were also able to increase cAMP response element-binding protein phosphorylation in neuronal nuclei in a glutamate receptor and calcium/calmodulin kinase-dependent manner. Moreover, the effect of ascorbate is not blocked by sulfinpyrazone and then does not depend on its uptake by retinal cells. Overall, these data indicate a novel molecular and functional target for vitamin C impacting on glutamate signaling in retinal neurons.


Subject(s)
Ascorbic Acid/pharmacology , Glutamates/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Retina/drug effects , Retina/metabolism , Vitamins/pharmacology , Animals , Biotinylation , Cells, Cultured , Chick Embryo , Chickens , Excitatory Amino Acid Transporter 3/metabolism , Extracellular Space/drug effects , Extracellular Space/metabolism , Signal Transduction/drug effects
7.
Sci Rep ; 7: 40912, 2017 01 18.
Article in English | MEDLINE | ID: mdl-28098256

ABSTRACT

Dopamine and glutamate are critical neurotransmitters involved in light-induced synaptic activity in the retina. In brain neurons, dopamine D1 receptors (D1Rs) and the cytosolic protein tyrosine kinase Src can, independently, modulate the behavior of NMDA-type glutamate receptors (NMDARs). Here we studied the interplay between D1Rs, Src and NMDARs in retinal neurons. We reveal that dopamine-mediated D1R stimulation provoked NMDAR hypofunction in retinal neurons by attenuating NMDA-gated currents, by preventing NMDA-elicited calcium mobilization and by decreasing the phosphorylation of NMDAR subunit GluN2B. This dopamine effect was dependent on upregulation of the canonical D1R/adenylyl cyclase/cAMP/PKA pathway, of PKA-induced activation of C-terminal Src kinase (Csk) and of Src inhibition. Accordingly, knocking down Csk or overexpressing a Csk phosphoresistant Src mutant abrogated the dopamine-induced NMDAR hypofunction. Overall, the interplay between dopamine and NMDAR hypofunction, through the D1R/Csk/Src/GluN2B pathway, might impact on light-regulated synaptic activity in retinal neurons.


Subject(s)
Dopamine/pharmacology , Receptors, Dopamine D1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Retina/drug effects , src-Family Kinases/metabolism , Animals , CSK Tyrosine-Protein Kinase , Calcium/metabolism , Chick Embryo , Chickens , Colforsin/pharmacology , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Dopamine D1/antagonists & inhibitors , Receptors, Dopamine D1/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Retina/metabolism , Signal Transduction/drug effects , Up-Regulation/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
8.
Glia ; 63(3): 497-511, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25421817

ABSTRACT

Microglial cells are the resident macrophages of the central nervous system. Their function is essential for neuronal tissue homeostasis. After inflammatory stimuli, microglial cells become activated changing from a resting and highly ramified cell shape to an amoeboid-like morphology. These morphological changes are associated with the release of proinflammatory cytokines and glutamate, as well as with high phagocytic activity. The acquisition of such phenotype has been associated with activation of cytoplasmic tyrosine kinases, including those of the Src family (SFKs). In this study, using both in vivo and in vitro inflammation models coupled to FRET-based time-lapse microscopy, lentiviruses-mediated shRNA delivery and genetic gain-of-function experiments, we demonstrate that among SFKs c-Src function is necessary and sufficient for triggering microglia proinflammatory signature, glutamate release, microglia-induced neuronal loss, and phagocytosis. c-Src inhibition in retinal neuroinflammation experimental paradigms consisting of intravitreal injection of LPS or ischemia-reperfusion injury significantly reduced microglia activation changing their morphology to a more resting phenotype and prevented neuronal apoptosis. Our data demonstrate an essential role for c-Src in microglial cell activation.


Subject(s)
Microglia/enzymology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Animals , Apoptosis/physiology , CSK Tyrosine-Protein Kinase , Cell Line , Cells, Cultured , Chickens , Gliosis/enzymology , Gliosis/pathology , Glutamic Acid/metabolism , HEK293 Cells , Humans , Inflammation/enzymology , Inflammation/pathology , Ischemia/enzymology , Ischemia/pathology , Lipopolysaccharides , Male , Mice , Microglia/pathology , Neurons/physiology , Phagocytosis/physiology , Rats, Wistar , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Retinal Neurons/pathology , Retinal Neurons/physiology , Tumor Necrosis Factor-alpha/metabolism , src-Family Kinases/metabolism
9.
Free Radic Biol Med ; 79: 45-55, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25486178

ABSTRACT

3-O-caffeoylquinic acid (3-CQA) is an isomer of chlorogenic acid, which has been shown to regulate lipopolysaccharide-induced tumor necrosis factor production in microglia. Whereas overactivation of microglia is associated with neuronal loss in brain diseases via reactive oxygen species (ROS) production and glutamate excitotoxicity, naïve (nonactivated) microglia are believed to generate little ROS under basal conditions, contributing to the modulation of synaptic activity and nerve tissue repair. However, the signaling pathways controlling basal ROS homeostasis in microglial cells are still poorly understood. Here we used time-lapse microscopy coupled with highly sensitive FRET biosensors (for detecting c-Src activation, ROS generation, and glutamate release) and lentivirus-mediated shRNA delivery to study the pathways involved in antioxidant-regulated ROS generation and how this associates with microglia-induced neuronal cell death. We report that 3-CQA abrogates the acquisition of an amoeboid morphology in microglia triggered by Aß oligomers or the HIV Tat peptide. Moreover, 3-CQA deactivates c-Src tyrosine kinase and abrogates c-Src activation during proinflammatory microglia stimulation, which shuts off ROS production in these cells. Moreover, forced increment of c-Src catalytic activity by overexpressing an inducible c-Src heteromerization construct in microglia increases ROS production, abrogating the 3-CQA effects. Whereas oxidant (hydrogen peroxide) stimulation dramatically enhances glutamate release from microglia, such release is diminished by the 3-CQA inhibition of c-Src/ROS generation, significantly alleviating cell death in cultures from embryonic neurons. Overall, we provide further mechanistic insight into the modulation of ROS production in cortical microglia, indicating antioxidant-regulated c-Src function as a pathway for controlling microglia-triggered oxidative damage.


Subject(s)
Chlorogenic Acid/pharmacology , Glutamic Acid/metabolism , Microglia/drug effects , Neurons/drug effects , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Animals , HEK293 Cells , Humans , Microglia/metabolism , Neurons/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Rats , Rats, Wistar
10.
Vitam Horm ; 96: 79-125, 2014.
Article in English | MEDLINE | ID: mdl-25189385

ABSTRACT

Nitric oxide (NO) is a very reactive molecule, and its short half-life would make it virtually invisible until its discovery. NO activates soluble guanylyl cyclase (sGC), increasing 3',5'-cyclic guanosine monophosphate levels to activate PKGs. Although NO triggers several phosphorylation cascades due to its ability to react with Fe II in heme-containing proteins such as sGC, it also promotes a selective posttranslational modification in cysteine residues by S-nitrosylation, impacting on protein function, stability, and allocation. In the central nervous system (CNS), NO synthesis usually requires a functional coupling of nitric oxide synthase I (NOS I) and proteins such as NMDA receptors or carboxyl-terminal PDZ ligand of NOS (CAPON), which is critical for specificity and triggering of selected pathways. NO also modulates CREB (cAMP-responsive element-binding protein), ERK, AKT, and Src, with important implications for nerve cell survival and differentiation. Differences in the regulation of neuronal death or survival by NO may be explained by several mechanisms involving localization of NOS isoforms, amount of NO being produced or protein sets being modulated. A number of studies show that NO regulates neurotransmitter release and different aspects of synaptic dynamics, such as differentiation of synaptic specializations, microtubule dynamics, architecture of synaptic protein organization, and modulation of synaptic efficacy. NO has also been associated with synaptogenesis or synapse elimination, and it is required for long-term synaptic modifications taking place in axons or dendrites. In spite of tremendous advances in the knowledge of NO biological effects, a full description of its role in the CNS is far from being completely elucidated.


Subject(s)
Central Nervous System/physiology , Nitric Oxide/metabolism , Signal Transduction/physiology , Animals , Central Nervous System/growth & development , Neuronal Plasticity/physiology , Neurons/physiology , Neurotransmitter Agents/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...