Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Biol Toxicol ; 39(5): 2089-2111, 2023 10.
Article in English | MEDLINE | ID: mdl-35137321

ABSTRACT

Increasing evidence from animal and epidemiological studies indicates that perinatal exposure to pesticides cause developmental neurotoxicity and may increase the risk for psychiatric disorders such as autism and intellectual disability. However, the underlying pathogenic mechanisms remain largely elusive. This work was aimed at testing the hypothesis that developmental exposure to different classes of pesticides hijacks intracellular neuronal signaling contributing to synaptic and behavioral alterations associated with neurodevelopmental disorders (NDD). Low concentrations of organochlorine (dieldrin, endosulfan, and chlordane) and organophosphate (chlorpyrifos and its oxon metabolite) pesticides were chronically dosed ex vivo (organotypic rat hippocampal slices) or in vivo (perinatal exposure in rats), and then biochemical, electrophysiological, behavioral, and proteomic studies were performed. All the pesticides tested caused prolonged activation of MAPK/ERK pathway in a concentration-dependent manner. Additionally, some of them impaired metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD). In the case of the pesticide chlordane, the effect was attributed to chronic modulation of MAPK/ERK signaling. These synaptic alterations were reproduced following developmental in vivo exposure to chlordane and chlorpyrifos-oxon, and were also associated with prototypical behavioral phenotypes of NDD, including impaired motor development, increased anxiety, and social and memory deficits. Lastly, proteomic analysis revealed that these pesticides differentially regulate the expression of proteins in the hippocampus with pivotal roles in brain development and synaptic signaling, some of which are associated with NDD. Based on these results, we propose a novel mechanism of synaptic dysfunction, involving chronic overactivation of MAPK and impaired mGluR-LTD, shared by different pesticides which may have important implications for NDD.


Subject(s)
Chlorpyrifos , Neurodevelopmental Disorders , Pesticides , Humans , Female , Pregnancy , Rats , Animals , Pesticides/toxicity , Chlorpyrifos/toxicity , Chlorpyrifos/metabolism , Chlordan/metabolism , Chlordan/pharmacology , Proteomics , Hippocampus/metabolism , Neuronal Plasticity , Neurodevelopmental Disorders/chemically induced , Neurodevelopmental Disorders/metabolism
2.
Sci Adv ; 8(47): eabq8109, 2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36417513

ABSTRACT

Neuronal connectivity and activity-dependent synaptic plasticity are fundamental properties that support brain function and cognitive performance. Phosphatidylinositol 3-kinase (PI3K) intracellular signaling controls multiple mechanisms mediating neuronal growth, synaptic structure, and plasticity. However, it is still unclear how these pleiotropic functions are integrated at molecular and cellular levels. To address this issue, we used neuron-specific virally delivered Cre expression to delete either p110α or p110ß (the two major catalytic isoforms of type I PI3K) from the hippocampus of adult mice. We found that dendritic and postsynaptic structures are almost exclusively supported by p110α activity, whereas p110ß controls neurotransmitter release and metabotropic glutamate receptor-dependent long-term depression at the presynaptic terminal. In addition to these separate functions, p110α and p110ß jointly contribute to N-methyl-d-aspartate receptor-dependent postsynaptic long-term potentiation. This molecular and functional specialization is reflected in different proteomes controlled by each isoform and in distinct behavioral alterations for learning/memory and sociability in mice lacking p110α or p110ß.

3.
Elife ; 102021 11 17.
Article in English | MEDLINE | ID: mdl-34787081

ABSTRACT

De novo protein synthesis is required for synapse modifications underlying stable memory encoding. Yet neurons are highly compartmentalized cells and how protein synthesis can be regulated at the synapse level is unknown. Here, we characterize neuronal signaling complexes formed by the postsynaptic scaffold GIT1, the mechanistic target of rapamycin (mTOR) kinase, and Raptor that couple synaptic stimuli to mTOR-dependent protein synthesis; and identify NMDA receptors containing GluN3A subunits as key negative regulators of GIT1 binding to mTOR. Disruption of GIT1/mTOR complexes by enhancing GluN3A expression or silencing GIT1 inhibits synaptic mTOR activation and restricts the mTOR-dependent translation of specific activity-regulated mRNAs. Conversely, GluN3A removal enables complex formation, potentiates mTOR-dependent protein synthesis, and facilitates the consolidation of associative and spatial memories in mice. The memory enhancement becomes evident with light or spaced training, can be achieved by selectively deleting GluN3A from excitatory neurons during adulthood, and does not compromise other aspects of cognition such as memory flexibility or extinction. Our findings provide mechanistic insight into synaptic translational control and reveal a potentially selective target for cognitive enhancement.


Subject(s)
Memory/physiology , Protein Biosynthesis/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Female , Male , Mechanistic Target of Rapamycin Complex 1 , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction
4.
Front Mol Neurosci ; 11: 380, 2018.
Article in English | MEDLINE | ID: mdl-30374290

ABSTRACT

Glycine receptors (GlyRs) containing the α2 subunit are highly expressed in the developing brain, where they regulate neuronal migration and maturation, promote spontaneous network activity and subsequent development of synaptic connections. Mutations in GLRA2 are associated with autism spectrum disorder, but the underlying pathophysiology is not described yet. Here, using Glra2-knockout mice, we found a GlyR-dependent effect on neonatal spontaneous activity of dorsal striatum medium spiny neurons (MSNs) and maturation of the incoming glutamatergic innervation. Our data demonstrate that functional GlyRs are highly expressed in MSNs of one-week-old mice, but they do not generate endogenous chloride-mediated tonic or phasic current. Despite of that, knocking out the Glra2 severely affects the shape of action potentials and impairs spontaneous activity and the frequency of miniature AMPA receptor-mediated currents in MSNs. This reduction in spontaneous activity and glutamatergic signaling can attribute to the observed changes in neonatal behavioral phenotypes as seen in ultrasonic vocalizations and righting reflex. In adult Glra2-knockout animals, the glutamatergic synapses in MSNs remain functionally underdeveloped. The number of glutamatergic synapses and release probability at presynaptic site remain unaffected, but the amount of postsynaptic AMPA receptors is decreased. This deficit is a consequence of impaired development of the neuronal circuitry since acute inhibition of GlyRs by strychnine in adult MSNs does not affect the properties of glutamatergic synapses. Altogether, these results demonstrate that GlyR-mediated signaling supports neonatal spontaneous MSN activity and, in consequence, promotes the functional maturation of glutamatergic synapses on MSNs. The described mechanism might shed light on the pathophysiological mechanisms in GLRA2-linked autism spectrum disorder cases.

5.
Nat Commun ; 8(1): 293, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819097

ABSTRACT

The brain cytoplasmic (BC1) RNA is a non-coding RNA (ncRNA) involved in neuronal translational control. Absence of BC1 is associated with altered glutamatergic transmission and maladaptive behavior. Here, we show that pyramidal neurons in the barrel cortex of BC1 knock out (KO) mice display larger excitatory postsynaptic currents and increased spontaneous activity in vivo. Furthermore, BC1 KO mice have enlarged spine heads and postsynaptic densities and increased synaptic levels of glutamate receptors and PSD-95. Of note, BC1 KO mice show aberrant structural plasticity in response to whisker deprivation, impaired texture novel object recognition and altered social behavior. Thus, our study highlights a role for BC1 RNA in experience-dependent plasticity and learning in the mammalian adult neocortex, and provides insight into the function of brain ncRNAs regulating synaptic transmission, plasticity and behavior, with potential relevance in the context of intellectual disabilities and psychiatric disorders.Brain cytoplasmic (BC1) RNA is a non-coding RNA that has been implicated in translational regulation, seizure, and anxiety. Here, the authors show that in the cortex, BC1 RNA is required for sensory deprivation-induced structural plasticity of dendritic spines, as well as for correct sensory learning and social behaviors.


Subject(s)
Learning/physiology , Neocortex/physiology , Neuronal Plasticity/physiology , Pyramidal Cells/physiology , RNA, Small Cytoplasmic/genetics , Animals , Base Sequence , Cells, Cultured , Dendritic Spines/metabolism , Dendritic Spines/physiology , Excitatory Postsynaptic Potentials/genetics , Excitatory Postsynaptic Potentials/physiology , In Situ Hybridization, Fluorescence , Male , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Neocortex/cytology , Neocortex/metabolism , Neuronal Plasticity/genetics , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Sensory Deprivation/physiology , Sequence Homology, Nucleic Acid , Social Behavior , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Vibrissae/metabolism , Vibrissae/physiology
6.
Sci Rep ; 7: 42788, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28202907

ABSTRACT

Recent studies indicate that calpain-1 is required for the induction of long-term potentiation (LTP) elicited by theta-burst stimulation in field CA1 of hippocampus. Here we determined the contribution of calpain-1 in another type of synaptic plasticity, the long-term depression (LTD) elicited by activation of type-I metabotropic glutamate receptors (mGluR-LTD). mGluR-LTD was associated with calpain-1 activation following T-type calcium channel opening, and resulted in the truncation of a regulatory subunit of PP2A, B56α. This signaling pathway was required for both the early and late phase of Arc translation during mGluR-LTD, through a mechanism involving mTOR and ribosomal protein S6 activation. In contrast, in hippocampal slices from calpain-1 knock-out (KO) mice, application of the mGluR agonist, DHPG, did not result in B56α truncation, increased Arc synthesis and reduced levels of membrane GluA1-containing AMPA receptors. Consistently, mGluR-LTD was impaired in calpain-1 KO mice, and the impairment could be rescued by phosphatase inhibitors, which also restored Arc translation in response to DHPG. Furthermore, calpain-1 KO mice exhibited impairment in fear memory extinction to tone presentation. These results indicate that calpain-1 plays a critical role in mGluR-LTD and is involved in many forms of synaptic plasticity and learning and memory.


Subject(s)
Calpain/genetics , Extinction, Psychological , Fear , Long-Term Synaptic Depression , Memory , Receptors, Metabotropic Glutamate/metabolism , Animals , Calcium Channels, T-Type/metabolism , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Protein Phosphatase 2/metabolism , Receptors, AMPA/metabolism
7.
Neuroscientist ; 23(3): 221-231, 2017 06.
Article in English | MEDLINE | ID: mdl-27188456

ABSTRACT

Although calpain was proposed to participate in synaptic plasticity and learning and memory more than 30 years ago, the mechanisms underlying its activation and the roles of different substrates have remained elusive. Recent findings have provided evidence that the two major calpain isoforms in the brain, calpain-1 and calpain-2, play opposite functions in synaptic plasticity. In particular, while calpain-1 activation is the initial trigger for certain forms of synaptic plasticity, that is, long-term potentiation, calpain-2 activation restricts the extent of plasticity. Moreover, while calpain-1 rapidly cleaves regulatory and cytoskeletal proteins, calpain-2-mediated stimulation of local protein synthesis reestablishes protein homeostasis. These findings have important implications for our understanding of learning and memory and disorders associated with impairment in these processes.


Subject(s)
Calpain/metabolism , Neuronal Plasticity/physiology , Animals , Brain/metabolism
8.
J Cell Biol ; 210(7): 1225-37, 2015 Sep 28.
Article in English | MEDLINE | ID: mdl-26391661

ABSTRACT

Estrogen is an important modulator of hippocampal synaptic plasticity and memory consolidation through its rapid action on membrane-associated receptors. Here, we found that both estradiol and the G-protein-coupled estrogen receptor 1 (GPER1) specific agonist G1 rapidly induce brain-derived neurotrophic factor (BDNF) release, leading to transient stimulation of activity-regulated cytoskeleton-associated (Arc) protein translation and GluA1-containing AMPA receptor internalization in field CA3 of hippocampus. We also show that type-I metabotropic glutamate receptor (mGluR) activation does not induce Arc translation nor long-term depression (LTD) at the mossy fiber pathway, as opposed to its effects in CA1, and it only triggers LTD after GPER1 stimulation. Furthermore, this form of mGluR-dependent LTD is associated with ubiquitination and proteasome-mediated degradation of GluA1, and is prevented by proteasome inhibition. Overall, our study identifies a novel mechanism by which estrogen and BDNF regulate hippocampal synaptic plasticity in the adult brain.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , CA3 Region, Hippocampal/metabolism , Neuronal Plasticity/physiology , Receptors, G-Protein-Coupled/metabolism , Animals , CA3 Region, Hippocampal/cytology , Male , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Receptors, Metabotropic Glutamate/metabolism
9.
J Neurosci ; 35(5): 2269-82, 2015 Feb 04.
Article in English | MEDLINE | ID: mdl-25653381

ABSTRACT

Dendritic protein synthesis and actin cytoskeleton reorganization are important events required for the consolidation of hippocampal LTP and memory. However, the temporal and spatial relationships between these two processes remain unclear. Here, we report that treatment of adult rat hippocampal slices with BDNF or with tetraethylammonium (TEA), which induces a chemical form of LTP, produces a rapid and transient increase in RhoA protein levels. Changes in RhoA were restricted to dendritic spines of CA3 and CA1 and require de novo protein synthesis regulated by mammalian target of rapamycin (mTOR). BDNF-mediated stimulation of RhoA activity, cofilin phosphorylation, and actin polymerization were completely suppressed by protein synthesis inhibitors. Furthermore, intrahippocampal injections of RhoA antisense oligodeoxynucleotides inhibited theta burst stimulation (TBS)-induced RhoA upregulation in dendritic spines and prevented LTP consolidation. Addition of calpain inhibitors after BDNF or TEA treatment maintained RhoA levels elevated and prolonged the effects of BDNF and TEA on actin polymerization. Finally, the use of isoform-selective calpain inhibitors revealed that calpain-2 was involved in RhoA synthesis, whereas calpain-1 mediated RhoA degradation. Overall, this mechanism provides a novel link between dendritic protein synthesis and reorganization of the actin cytoskeleton in hippocampal dendritic spines during LTP consolidation.


Subject(s)
CA1 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/metabolism , Long-Term Potentiation , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton/metabolism , Actin Depolymerizing Factors/metabolism , Actins/metabolism , Animals , Brain-Derived Neurotrophic Factor/pharmacology , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/physiology , Calpain/antagonists & inhibitors , Calpain/metabolism , Dendritic Spines/metabolism , Male , Organ Specificity , Phosphorylation , Protein Synthesis Inhibitors/pharmacology , Proteolysis , Rats , Rats, Sprague-Dawley , Tetraethylammonium/pharmacology , rhoA GTP-Binding Protein/genetics
10.
Brain Res ; 1621: 73-81, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-25482663

ABSTRACT

Since its discovery by Bliss and Lomo, the phenomenon of long-term potentiation (LTP) has been extensively studied, as it was viewed as a potential cellular mechanism of learning and memory. Over the years, many signaling cascades have been implicated in its induction, consolidation and maintenance, raising questions regarding its real significance. Here, we review several of the most commonly studie signaling cascades and discuss how they converge on a common set of mechanisms likely to be involved in the maintenance of LTP. We further argue that the existence of cross-talks between these different signaling cascades can not only account for several discrepancies in the literature, but also account for the existence of different forms of LTP, which can be engaged by different types of stimulus parameters under different experimental conditions. Finally, we discuss how the understanding of the diversity of LTP mechanisms can help us understand the diversity of the types of learning and memory. This article is part of a Special Issue entitled SI: Brain and Memory.


Subject(s)
Hippocampus/metabolism , Learning/physiology , Long-Term Potentiation , Memory/physiology , Signal Transduction , Synapses/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calpain/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dendritic Spines/metabolism , Humans , MAP Kinase Signaling System , Neurons/metabolism
11.
Article in English | MEDLINE | ID: mdl-24611062

ABSTRACT

Estrogen rapidly modulates hippocampal synaptic plasticity by activating selective membrane-associated receptors. Reorganization of the actin cytoskeleton and stimulation of mammalian target of rapamycin (mTOR)-mediated protein synthesis are two major events required for the consolidation of hippocampal long-term potentiation and memory. Estradiol regulates synaptic plasticity by interacting with both processes, but the underlying molecular mechanisms are not yet fully understood. Here, we used acute rat hippocampal slices to analyze the mechanisms underlying rapid changes in mTOR activity and actin polymerization elicited by estradiol. Estradiol-induced mTOR phosphorylation was preceded by rapid and transient activation of both extracellular signal-regulated kinase (ERK) and protein kinase B (Akt) and by phosphatase and tensin homolog (PTEN) degradation. These effects were prevented by calpain and ERK inhibitors. Estradiol-induced mTOR stimulation did not require activation of classical estrogen receptors (ER), as specific ERα and ERß agonists (PPT and DPN, respectively) failed to mimic this effect, and ER antagonists could not block it. Estradiol rapidly activated both RhoA and p21-activated kinase (PAK). Furthermore, a specific inhibitor of RhoA kinase (ROCK), H1152, and a potent and specific PAK inhibitor, PF-3758309, blocked estradiol-induced cofilin phosphorylation and actin polymerization. ER antagonists also blocked these effects of estrogen. Consistently, both PPT and DPN stimulated PAK and cofilin phosphorylation as well as actin polymerization. Finally, the effects of estradiol on actin polymerization were insensitive to protein synthesis inhibitors, but its stimulation of mTOR activity was impaired by latrunculin A, a drug that disrupts actin filaments. Taken together, our results indicate that estradiol regulates local protein synthesis and cytoskeletal reorganization via different molecular mechanisms and signaling pathways.

12.
Nat Commun ; 5: 3051, 2014.
Article in English | MEDLINE | ID: mdl-24394804

ABSTRACT

Overexpression of suprachiasmatic nucleus circadian oscillatory protein (SCOP), a negative ERK regulator, blocks long-term memory encoding. Inhibition of calpain-mediated SCOP degradation also prevents the formation of long-term memory, suggesting rapid SCOP breakdown is necessary for memory encoding. However, whether SCOP levels also control the magnitude of long-term synaptic plasticity is unknown. Here we show that following synaptic activity-induced SCOP degradation, SCOP is rapidly replaced via mTOR-mediated protein synthesis. We further show that early SCOP degradation is specifically catalysed by µ-calpain, whereas late SCOP resynthesis is mediated by m-calpain. We propose that µ-calpain promotes long-term potentiation induction by degrading SCOP and activating ERK, whereas m-calpain activation limits the magnitude of potentiation by terminating the ERK response via enhanced SCOP synthesis. This unique braking mechanism could account for the advantages of spaced versus massed training in the formation of long-term memory.


Subject(s)
Hippocampus/physiology , Long-Term Potentiation/physiology , Memory, Long-Term/physiology , Nuclear Proteins/physiology , Animals , Calpain/physiology , MAP Kinase Signaling System/physiology , Male , Neuronal Plasticity/physiology , Phosphoprotein Phosphatases , Rats , Rats, Sprague-Dawley
13.
J Neurosci ; 33(48): 18880-92, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24285894

ABSTRACT

Prolonged calpain activation is widely recognized as a key component of neurodegeneration in a variety of pathological conditions. Numerous reports have also indicated that synaptic activation of NMDA receptors (NMDARs) provides neuroprotection against a variety of insults. Here, we report the paradoxical finding that such neuroprotection involves calpain activation. NMDAR activation in cultured rat cortical neurons was neuroprotective against starvation and oxidative stress-induced damage. It also resulted in the degradation of two splice variants of PH domain and Leucine-rich repeat Protein Phosphatase 1 (PHLPP1), PHLPP1α and PHLPP1ß, which inhibit the Akt and ERK1/2 pathways. Synaptic NMDAR-induced neuroprotection and PHLPP1 degradation were blocked by calpain inhibition. Lentiviral knockdown of PHLPP1 mimicked the neuroprotective effects of synaptic NMDAR activation and occluded the effects of calpain inhibition on neuroprotection. In contrast to synaptic NMDAR activation, extrasynaptic NMDAR activation had no effect on PHLPP1 and the Akt and ERK1/2 pathways, but resulted in calpain-mediated degradation of striatal-enriched protein tyrosine phosphatase (STEP) and neuronal death. Using µ-calpain- and m-calpain-selective inhibitors and µ-calpain and m-calpain siRNAs, we found that µ-calpain-dependent PHLPP1 cleavage was involved in synaptic NMDAR-mediated neuroprotection, while m-calpain-mediated STEP degradation was associated with extrasynaptic NMDAR-induced neurotoxicity. Furthermore, m-calpain inhibition reduced while µ-calpain knockout exacerbated NMDA-induced neurotoxicity in acute mouse hippocampal slices. Thus, synaptic NMDAR-coupled µ-calpain activation is neuroprotective, while extrasynaptic NMDAR-coupled m-calpain activation is neurodegenerative. These results help to reconcile a number of contradictory results in the literature and have critical implications for the understanding and potential treatment of neurodegenerative diseases.


Subject(s)
Calpain/physiology , Nerve Degeneration/physiopathology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/physiology , Animals , Calpain/antagonists & inhibitors , Calpain/genetics , Cell Death/physiology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Immunohistochemistry , Immunoprecipitation , Lentivirus/genetics , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Knockout , Nuclear Proteins/physiology , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/physiology , Primary Cell Culture , Protein Tyrosine Phosphatases/metabolism , RNA, Small Interfering , Rats , Receptors, N-Methyl-D-Aspartate/agonists
14.
J Neurosci ; 33(10): 4317-28, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23467348

ABSTRACT

Memory consolidation has been suggested to be protein synthesis dependent. Previous data indicate that BDNF-induced dendritic protein synthesis is a key event in memory formation through activation of the mammalian target of rapamycin (mTOR) pathway. BDNF also activates calpain, a calcium-dependent cysteine protease, which has been shown to play a critical role in learning and memory. This study was therefore directed at testing the hypothesis that calpain activity is required for BDNF-stimulated local protein synthesis, and at identifying the underlying molecular mechanism. In rat hippocampal slices, cortical synaptoneurosomes, and cultured neurons, BDNF-induced mTOR pathway activation and protein translation were blocked by calpain inhibition. BDNF treatment rapidly reduced levels of hamartin and tuberin, negative regulators of mTOR, in a calpain-dependent manner. Treatment of brain homogenates with purified calpain-1 and calpain-2 truncated both proteins. BDNF treatment increased phosphorylation of both Akt and ERK, but only the effect on Akt was blocked by calpain inhibition. Levels of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), a phosphatase that inactivates Akt, were decreased following BDNF treatment, and calpain inhibition reversed this effect. Calpain-2, but not calpain-1, treatment of brain homogenates resulted in PTEN degradation. In cultured cortical neurons, knockdown of calpain-2, but not calpain-1, by small interfering RNA completely suppressed the effect of BDNF on mTOR activation. Our results reveal a critical role for calpain-2 in BDNF-induced mTOR signaling and dendritic protein synthesis via PTEN, hamartin, and tuberin degradation. This mechanism therefore provides a link between proteolysis and protein synthesis that might contribute to synaptic plasticity.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Calpain/metabolism , Dendrites/drug effects , Dendrites/metabolism , Neurons/ultrastructure , PTEN Phosphohydrolase/metabolism , Analysis of Variance , Animals , Butadienes/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calpain/genetics , Cells, Cultured , Cerebral Cortex/cytology , Dendrites/ultrastructure , Dipeptides/pharmacology , Embryo, Mammalian , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , Hippocampus/cytology , In Vitro Techniques , Male , Neurons/drug effects , Nitriles/pharmacology , Oxazines/pharmacology , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Synaptosomes/drug effects , Synaptosomes/metabolism , TOR Serine-Threonine Kinases/metabolism , Transfection , Tuberous Sclerosis Complex 1 Protein , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
15.
Endocrinology ; 153(2): 847-60, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22166974

ABSTRACT

Dieldrin is an endocrine disruptor that accumulates in mammalian adipose tissue and brain. It induces convulsions due to its antagonism of the γ-aminobutyric acid A receptor (GABA(A)R). We have previously reported that long-term exposure to dieldrin causes the internalization of the N-methyl-D-aspartate receptor (NMDAR) as a result of persistent GABA(A)R inhibition. Because the neurosteroids 17ß-estradiol (E2) and allopregnanolone are known to modulate the function and trafficking of GABA(A)R and NMDAR, we examined the effects of E2 and allopregnanolone on dieldrin-induced GABA(A)R inhibition, NMDAR internalization, and neuronal death in cortical neurons. We found that 1 nM E2 increased the membrane expression of NR1/NR2B receptors and postsynaptic density 95 but did not induce their physical association. In contrast, 10 nM E2 had no effect on these proteins but reduced NR2A membrane expression. We also found that exposure to 60 nM dieldrin for 6 d in vitro caused the internalization of NR1 and NR2B but not NR2A. Treatment with either 1 nM E2 or 10 µM allopregnanolone prevented the dieldrin-induced reduction in membrane levels of the NR1/NR2B receptors. Furthermore, prolonged exposure to 200 nM dieldrin down-regulated the expression of NR2A; this was inhibited only by allopregnanolone. Although both hormones restored NMDAR function, as measured by the NMDA-induced rise in intracellular calcium, allopregnanolone (but not E2) reversed the inhibition of GABA(A)R and neuronal death caused by prolonged exposure to dieldrin. Our results indicate that allopregnanolone protects cortical neurons against the neurotoxicity caused by long-term exposure to dieldrin by maintaining GABA(A)R and NMDAR functionality.


Subject(s)
Dieldrin/adverse effects , Neurons/drug effects , Pregnanolone/pharmacology , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Anesthetics/pharmacology , Animals , Cell Membrane , Cerebral Cortex/cytology , Disks Large Homolog 4 Protein , Embryo, Mammalian/cytology , Endocrine Disruptors/adverse effects , Estradiol/pharmacology , Female , Gene Expression Regulation/physiology , Glutamic Acid/metabolism , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neurons/metabolism , Pregnancy , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Synaptic Transmission/physiology
16.
Methods Mol Biol ; 758: 253-65, 2011.
Article in English | MEDLINE | ID: mdl-21815071

ABSTRACT

Glutamate is the mayor excitatory neurotransmitter in vertebrate nervous system. It has a crucial role in most brain functions under physiological conditions through the activation of both ionotropic and metabotropic glutamate receptors. In addition, extracellular glutamate concentration is tightly regulated through different excitatory amino acid transporters (EAAT). Glutamate neurotransmission is also involved in the neurotoxic effects of many environmental chemicals and drugs. Furthermore, homeostatic changes in glutamate neurotransmission appear in response to prolonged block/enhancement of electrical activity. Here, we describe different approaches to evaluate alterations in glutamate neurotransmission regarding glutamate receptors and glutamate transporters by using primary cultures of neurons and astrocytes. The methods are based on the increased fluorescence of calcium-sensitive probes in response to glutamate agonists, on radioligand binding to glutamate receptors and transport sites, and on inmunocytochemistry visualization of glutamate receptors.


Subject(s)
Glutamic Acid/pharmacology , Neurons/metabolism , Neurotransmitter Agents/metabolism , Primary Cell Culture , Amino Acid Transport System X-AG/metabolism , Aniline Compounds , Animals , Aspartic Acid/metabolism , Biological Transport, Active , Brain/cytology , Calcium Signaling , Dizocilpine Maleate/pharmacology , Embryo, Mammalian/cytology , Female , Fluorescent Dyes , Glutamic Acid/metabolism , Glutamic Acid/physiology , Homeostasis , Mice , Pregnancy , Radioligand Assay , Receptors, N-Methyl-D-Aspartate/agonists , Xanthenes
17.
Toxicol Sci ; 120(2): 413-27, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21278053

ABSTRACT

The organochlorine chemicals endosulfan, dieldrin, and γ-hexachlorocyclohexane (lindane) are persistent pesticides to which people are exposed mainly via diet. Their antagonism of the γ-aminobutyric acid-A (GABA(A)) receptor makes them convulsants. They are also endocrine disruptors because of their interaction with the estrogen receptor (ER). Here, we study the effects of dieldrin, endosulfan, and lindane on ERs in primary cultures of cortical neurons (CN) and cerebellar granule cells (CGC). All the compounds tested inhibited the binding of [(3)H]-estradiol to the ER in both CN and CGC, with dieldrin in CGC showing the highest affinity. We also determined the effects of the pesticides on protein kinase B (Akt) and extracellular-regulated kinase 1 and 2 (ERK1/2) phosphorylation. Dieldrin and endosulfan increased Akt phosphorylation in CN, which was inhibited by the ERß antagonist 4-[2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl]phenol. Instead, Akt and ERK1/2 phosphorylation induced by dieldrin in CGC was mediated by multiple activation of ERα, ERß, and G protein-coupled receptor 30. Lindane did not activate these pathways, but it inhibited estradiol-mediated Akt and ERK1/2 activation. In CN, all the chemicals activated ERK1/2 through a mechanism involving GABA(A) and glutamate receptors. Long-term exposure to these pesticides reduced the levels of ERα, but not of ERß. Moreover, extracts of CN treated with endosulfan, dieldrin, or lindane induced cell proliferation in MCF-7 human breast cancer-derived cells, whereas only extracts of CGC treated with dieldrin induced MCF-7 cell proliferation. Overall, the observed alterations on ER-mediated signaling and ER levels in neurons might contribute to the neurotoxicity of these organochlorine pesticides.


Subject(s)
Endocrine Disruptors/toxicity , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Hydrocarbons, Chlorinated/toxicity , Neurons/drug effects , Pesticides/toxicity , Animals , Animals, Newborn , Blotting, Western , Cell Culture Techniques , Cells, Cultured , Cerebellum/cytology , Cerebellum/embryology , Cerebellum/growth & development , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Dieldrin/chemistry , Dieldrin/toxicity , Endocrine Disruptors/chemistry , Endosulfan/chemistry , Endosulfan/toxicity , Extracellular Signal-Regulated MAP Kinases/metabolism , Hexachlorocyclohexane/chemistry , Hexachlorocyclohexane/toxicity , Humans , Hydrocarbons, Chlorinated/chemistry , Mice , Neurons/metabolism , Pesticides/chemistry , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , Receptors, GABA-A/metabolism
18.
Toxicol Sci ; 113(1): 138-49, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19812363

ABSTRACT

Dieldrin was previously used as a pesticide. Although its use has been discontinued, humans are still exposed to it due to its high environmental persistence and because it accumulates in the adipose tissue of animals. Acute exposure to dieldrin provokes convulsions due to its antagonism on the gamma-aminobutyric acid-A (GABA(A)) receptor. However, little is known about the effects of low chronic exposure to this pollutant. In the present work, we use primary cultures of cortical neurons to study the mechanisms involved in the toxic action of dieldrin. We found that 2 and 6 days in vitro (DIV) exposure to a subcytotoxic concentration (60nM) of dieldrin reduced the increase in intracellular calcium concentration ([Ca(2+)](i)) and the excitotoxicity caused by glutamate. Exposure to dieldrin for 6 DIV induced N-methyl-D-aspartate receptor (NMDAR) internalization and reduced metabotropic glutamate receptor 5 (mGLUR5) levels. Double immunostaining for NMDAR and mGLUR5 showed that these receptors lose colocalization on the cell membrane in neurons treated with dieldrin. No changes were observed in receptor functionalities or receptor levels after 2 DIV of exposure to dieldrin. However, the increase in [Ca(2+)](i) induced by coactivation of NMDAR and mGLUR5 was significantly reduced. Thus, a functional interaction between the two receptors seems to play an important role in glutamate-induced excitotoxicity. We confirm that permanent blockade of the GABA(A) receptor by this persistent pesticide triggers adaptive neuronal changes consisting of a reduction of glutamatergic neurotransmission. This might explain the cognitive and learning deficits observed in animals after chronic treatment with dieldrin.


Subject(s)
Cerebral Cortex/drug effects , Dieldrin/toxicity , Glutamic Acid/metabolism , Neurons/drug effects , Pesticides/toxicity , Receptors, Metabotropic Glutamate/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Synaptic Transmission/drug effects , Animals , Calcium/metabolism , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dose-Response Relationship, Drug , Down-Regulation , Female , GABA Antagonists/toxicity , GABA-A Receptor Antagonists , Gestational Age , L-Lactate Dehydrogenase/metabolism , Mice , Neurons/metabolism , Neurons/pathology , Pregnancy , Receptor, Metabotropic Glutamate 5 , Receptors, GABA-A/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL