Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Mol Psychiatry ; 29(5): 1322-1337, 2024 May.
Article in English | MEDLINE | ID: mdl-38233468

ABSTRACT

Fear-related pathologies are among the most prevalent psychiatric conditions, having inappropriate learned fear and resistance to extinction as cardinal features. Exposure therapy represents a promising therapeutic approach, the efficiency of which depends on inter-individual variation in fear extinction learning, which neurobiological basis is unknown. We characterized a model of extinction learning, whereby fear-conditioned mice were categorized as extinction (EXT)-success or EXT-failure, according to their inherent ability to extinguish fear. In the lateral amygdala, GluN2A-containing NMDAR are required for LTP and stabilization of fear memories, while GluN2B-containing NMDAR are required for LTD and fear extinction. EXT-success mice showed attenuated LTP, strong LTD and higher levels of synaptic GluN2B, while EXT-failure mice showed strong LTP, no LTD and higher levels of synaptic GluN2A. Neurotrophin 3 (NT3) infusion in the lateral amygdala was sufficient to rescue extinction deficits in EXT-failure mice. Mechanistically, activation of tropomyosin receptor kinase C (TrkC) with NT3 in EXT-failure slices attenuated lateral amygdala LTP, in a GluN2B-dependent manner. Conversely, blocking endogenous NT3-TrkC signaling with TrkC-Fc chimera in EXT-success slices strengthened lateral amygdala LTP. Our data support a key role for the NT3-TrkC system in inter-individual differences in fear extinction in rodents, through modulation of amygdalar NMDAR composition and synaptic plasticity.


Subject(s)
Amygdala , Extinction, Psychological , Fear , Individuality , Mice, Inbred C57BL , Neuronal Plasticity , Neurotrophin 3 , Receptor, trkC , Receptors, N-Methyl-D-Aspartate , Animals , Fear/physiology , Extinction, Psychological/physiology , Amygdala/metabolism , Amygdala/physiology , Mice , Neuronal Plasticity/physiology , Male , Receptors, N-Methyl-D-Aspartate/metabolism , Receptor, trkC/metabolism , Neurotrophin 3/metabolism , Long-Term Potentiation/physiology , Signal Transduction/physiology , Conditioning, Classical/physiology
2.
Cell Mol Life Sci ; 80(6): 173, 2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37266732

ABSTRACT

Mitochondria are present in the pre- and post-synaptic regions, providing the energy required for the activity of these very specialized neuronal compartments. Biogenesis of synaptic mitochondria takes place in the cell body, and these organelles are then transported to the synapse by motor proteins that carry their cargo along microtubule tracks. The transport of mitochondria along neurites is a highly regulated process, being modulated by the pattern of neuronal activity and by extracellular cues that interact with surface receptors. These signals act by controlling the distribution of mitochondria and by regulating their activity. Therefore, mitochondria activity at the synapse allows the integration of different signals and the organelles are important players in the response to synaptic stimulation. Herein we review the available evidence regarding the regulation of mitochondrial dynamics by neuronal activity and by neuromodulators, and how these changes in the activity of mitochondria affect synaptic communication.


Subject(s)
Mitochondria , Neurons , Mitochondria/metabolism , Neurons/metabolism , Organelles/metabolism , Kinesins/metabolism , Microtubules/metabolism
3.
Biochem Soc Trans ; 51(1): 315-330, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36629507

ABSTRACT

AMPA-type receptors for the neurotransmitter glutamate are very dynamic entities, and changes in their synaptic abundance underlie different forms of synaptic plasticity, including long-term synaptic potentiation (LTP), long-term depression (LTD) and homeostatic scaling. The different AMPA receptor subunits (GluA1-GluA4) share a common modular structure and membrane topology, and their intracellular C-terminus tail is responsible for the interaction with intracellular proteins important in receptor trafficking. The latter sequence differs between subunits and contains most sites for post-translational modifications of the receptors, including phosphorylation, O-GlcNAcylation, ubiquitination, acetylation, palmitoylation and nitrosylation, which affect differentially the various subunits. Considering that each single subunit may undergo modifications in multiple sites, and that AMPA receptors may be formed by the assembly of different subunits, this creates multiple layers of regulation of the receptors with impact in synaptic function and plasticity. This review discusses the diversity of mechanisms involved in the post-translational modification of AMPA receptor subunits, and their impact on the subcellular distribution and synaptic activity of the receptors.


Subject(s)
Receptors, AMPA , Synapses , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Synapses/metabolism , Neuronal Plasticity/physiology , Long-Term Potentiation/physiology , Protein Processing, Post-Translational
4.
J Neuroinflammation ; 19(1): 44, 2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35135578

ABSTRACT

BACKGROUND: Multiple sclerosis is an inflammatory and degenerative disease of the central nervous system (CNS) characterized by demyelination and concomitant axonal loss. The lack of a single specific test, and the similarity to other inflammatory diseases of the central nervous system, makes it difficult to have a clear diagnosis of multiple sclerosis. Therefore, laboratory tests that allows a clear and definite diagnosis, as well as to predict the different clinical courses of the disease are of utmost importance. Herein, we compared the cerebrospinal fluid (CSF) proteome of patients with multiple sclerosis (in the relapse-remitting phase of the disease) and other diseases of the CNS (inflammatory and non-inflammatory) aiming at identifying reliable biomarkers of multiple sclerosis. METHODS: CSF samples from the discovery group were resolved by 2D-gel electrophoresis followed by identification of the protein spots by mass spectrometry. The results were analyzed using univariate (Student's t test) and multivariate (Hierarchical Cluster Analysis, Principal Component Analysis, Linear Discriminant Analysis) statistical and numerical techniques, to identify a set of protein spots that were differentially expressed in CSF samples from patients with multiple sclerosis when compared with other two groups. Validation of the results was performed in samples from a different set of patients using quantitative (e.g., ELISA) and semi-quantitative (e.g., Western Blot) experimental approaches. RESULTS: Analysis of the 2D-gels showed 13 protein spots that were differentially expressed in the three groups of patients: Alpha-1-antichymotrypsin, Prostaglandin-H2-isomerase, Retinol binding protein 4, Transthyretin (TTR), Apolipoprotein E, Gelsolin, Angiotensinogen, Agrin, Serum albumin, Myosin-15, Apolipoprotein B-100 and EF-hand calcium-binding domain-containing protein. ELISA experiments allowed validating part of the results obtained in the proteomics analysis and showed that some of the alterations in the CSF proteome are also mirrored in serum samples from multiple sclerosis patients. CSF of multiple sclerosis patients was characterized by TTR oligomerization, thus highlighting the importance of analyzing posttranslational modifications of the proteome in the identification of novel biomarkers of the disease. CONCLUSIONS: The model built based on the results obtained upon analysis of the 2D-gels and in the validation phase attained an accuracy of about 80% in distinguishing multiple sclerosis patients and the other two groups.


Subject(s)
Multiple Sclerosis , Biomarkers/cerebrospinal fluid , Electrophoresis, Gel, Two-Dimensional , Humans , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/diagnosis , Protein Processing, Post-Translational , Proteome/analysis
5.
FASEB J ; 35(8): e21763, 2021 08.
Article in English | MEDLINE | ID: mdl-34245609

ABSTRACT

The synaptic expression of glutamate receptors of the α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) type is dynamically controlled by interaction with binding partners and auxiliary proteins. These proteins can be regulated by posttranslational modifications, including ubiquitination. In this work, we investigated the regulation of glutamate receptor interacting protein-associated protein 1 (GRASP1) by ubiquitin-dependent mechanisms and its impact on surface expression and activity of synaptic AMPA receptors. Cotransfection of GFP-ubiquitin decreased myc-GRASP1 protein levels in HEK293T cells, and this effect was inhibited upon transfection of an ubiquitin mutant that cannot be ubiquitinated on Lys48. In addition, transfection of cultured hippocampal neurons with GFP-ubiquitin reduced the dendritic levels of endogenous GRASP1 and decreased the surface expression of GluA1 AMPA receptor subunits, an effect that was partly reversed by cotransfection with GRASP1. Similarly, transfection of hippocampal neurons with GFP-ubiquitin decreased the amplitude of miniature excitatory postsynaptic currents (mEPSCs) mediated by Ca2+ -impermeable AMPA receptors, and this effect was abrogated by cotransfection of GRASP1. Together, the results show a role for ubiquitination in the regulation of the postsynaptic protein GRASP1, which has an impact on the surface distribution of AMPA receptors and on their activity at the synapse.


Subject(s)
Calcium Signaling , Gene Expression Regulation , Golgi Matrix Proteins/metabolism , Hippocampus/metabolism , Neurons/metabolism , Receptors, AMPA/biosynthesis , Ubiquitination , Animals , Golgi Matrix Proteins/genetics , HEK293 Cells , Humans , Rats , Receptors, AMPA/genetics
6.
Eur J Neurosci ; 52(1): 2771-2780, 2020 07.
Article in English | MEDLINE | ID: mdl-32168385

ABSTRACT

Previous studies about the modulation of the vasculature by CO were performed exclusively in male or sexually immature animals. Understanding the sex differences regarding systemic drug processing and pharmacodynamics is an important feature for safety assessment of drug dosing and efficacy. In this work, we used CORM-A1 as source of CO to examine the effects of this gasotransmitter on brain perfusion and the sex-dependent differences. Dynamic contrast-enhanced imaging (DCE)-based analysis was used to characterize the properties of CO in the modulation of cerebral vasculature in vivo, in adult C57BL/6 healthy mice. Perfusion of the temporal muscle, maxillary vein and in hippocampus, cortex and striatum was analysed for 108 min following CORM-A1 administration of 3 or 5 mg/kg. Under control conditions, brain perfusion was lower in females when compared with males. Under CO treatment, females showed a surprisingly overall reduced perfusion compared with controls (F = 3.452, p = .0004), while no major alterations (or even the expected increase) were observed in males. Cortical structures were only modulated in females. A striking female-dominated vasoconstriction effect was observed in the hippocampus and striatum following administration of CO, in this mixed-sex cohort. As these two regions are implicated in episodic and procedural memory formation, CO may have a relevant impact in learning and memory.


Subject(s)
Carbon Monoxide , Sex Characteristics , Animals , Female , Hippocampus , Male , Memory , Mice , Mice, Inbred C57BL
7.
J Neurosci ; 35(8): 3319-29, 2015 Feb 25.
Article in English | MEDLINE | ID: mdl-25716833

ABSTRACT

The neurotrophin brain-derived neurotrophic factor (BDNF) mediates activity-dependent long-term changes of synaptic strength in the CNS. The effects of BDNF are partly mediated by stimulation of local translation, with consequent alterations in the synaptic proteome. The ubiquitin-proteasome system (UPS) also plays an important role in protein homeostasis at the synapse by regulating synaptic activity. However, whether BDNF acts on the UPS to mediate the effects on long-term synaptic potentiation (LTP) has not been investigated. In the present study, we show similar and nonadditive effects of BDNF and proteasome inhibition on the early phase of synaptic potentiation (E-LTP) induced by theta-burst stimulation of rat hippocampal CA1 synapses. The effects of BDNF were blocked by the proteasome activator IU1, suggesting that the neurotrophin acts by decreasing proteasome activity. Accordingly, BDNF downregulated the proteasome activity in cultured hippocampal neurons and in hippocampal synaptoneurosomes. Furthermore, BDNF increased the activity of the deubiquitinating enzyme UchL1 in synaptoneurosomes and upregulated free ubiquitin. In contrast to the effects on posttetanic potentiation, proteasome activity was required for BDNF-mediated LTP. These results show a novel role for BDNF in UPS regulation at the synapse, which is likely to act together with the increased translation activity in the regulation of the synaptic proteome during E-LTP.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , CA1 Region, Hippocampal/metabolism , Long-Term Potentiation , Proteasome Endopeptidase Complex/metabolism , Animals , CA1 Region, Hippocampal/physiology , Cells, Cultured , Male , Proteasome Endopeptidase Complex/drug effects , Pyrroles/pharmacology , Pyrrolidines/pharmacology , Rats , Rats, Wistar , Synaptosomes/drug effects , Synaptosomes/metabolism , Theta Rhythm , Ubiquitin/metabolism , Ubiquitin Thiolesterase/metabolism
8.
J Neurochem ; 139(6): 918-920, 2016 12.
Article in English | MEDLINE | ID: mdl-27859239

ABSTRACT

This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".


Subject(s)
Brain Diseases/physiopathology , Congresses as Topic , Synapses/chemistry , Synapses/physiology , Animals , Brain Diseases/metabolism , Humans , Neurochemistry
9.
J Neurochem ; 139(6): 997-1018, 2016 12.
Article in English | MEDLINE | ID: mdl-27424566

ABSTRACT

Neuronal excitability depends on the balance between inhibitory and excitatory neurotransmission, which in the CNS are mainly mediated by GABA and glutamate respectively. The plasticity of glutamatergic synapses and the underlying molecular mechanisms have been characterized to a large extent. In comparison, much less is known regarding the plasticity of GABAergic synapses, which is also important in the maintenance of the excitatory/inhibitory balance. GABAergic synapses, similarly to the glutamatergic synapses, adjust their strength depending on the pattern of neuronal activity. These alterations take place in the pre- and postsynaptic compartments, and short- and long-term alterations have been described. At the postsynaptic level the plasticity of inhibitory synapses is largely mediated by modulation of the expression, localization and function of GABAA receptors, by mechanisms involving the participation of scaffold proteins and structural molecules. This review is focused on the key mechanisms that regulate GABAA receptor trafficking in response to alterations in neuronal activity or to stimulation of plasma membrane receptors. These alterations in GABAergic neurotransmission are important in the refinement of the pattern of activity of neuronal networks. In this work, we review some of the mechanisms contributing to the plasticity of inhibitory synapses in the CNS, focusing on the regulation of GABAA receptor (GABAA R) trafficking in response to alterations in neuronal activity or to stimulation of different classes of plasma membrane-associated receptors. Alterations in these mechanisms are important in the refinement of neuronal network activity. This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".


Subject(s)
GABAergic Neurons/metabolism , Neuronal Plasticity/physiology , Receptors, GABA-A/metabolism , Synapses/metabolism , Animals , Humans , Protein Transport/physiology , Synaptic Transmission/physiology
10.
Neurobiol Dis ; 89: 223-34, 2016 May.
Article in English | MEDLINE | ID: mdl-26581639

ABSTRACT

Global cerebral ischemia induces selective degeneration of specific subsets of neurons throughout the brain, particularly in the hippocampus and cortex. One of the major hallmarks of cerebral ischemia is excitotoxicity, characterized by overactivation of glutamate receptors leading to intracellular Ca(2+) overload and ultimately neuronal demise. N-methyl-d-aspartate receptors (NMDARs) are considered to be largely responsible for excitotoxic injury due to their high Ca(2+) permeability. In the hippocampus and cortex, these receptors are most prominently composed of combinations of two GluN1 subunits and two GluN2A and/or GluN2B subunits. Due to the controversy regarding the differential role of GluN2A and GluN2B subunits in excitotoxic cell death, we investigated the role of GluN2B in the activation of pro-death signaling following an in vitro model of global ischemia, oxygen and glucose deprivation (OGD). For this purpose, we used GluN2B(-/-) mouse cortical cultures and observed that OGD-induced damage was reduced in these neurons, and partially prevented in wild-type rat neurons by a selective GluN2B antagonist. Notably, we found a crucial role of the C-terminal domain of the GluN2B subunit in triggering excitotoxic signaling. Indeed, expression of YFP-GluN2B C-terminus mutants for the binding sites to post-synaptic density protein 95 (PSD95), Ca(2+)-calmodulin kinase IIα (CaMKIIα) or clathrin adaptor protein 2 (AP2) failed to mediate neuronal death in OGD conditions. We focused on the GluN2B-CaMKIIα interaction and found a determinant role of this interaction in OGD-induced death. Inhibition or knock-down of CaMKIIα exerted a neuroprotective effect against OGD-induced death, whereas overexpression of this kinase had a detrimental effect. Importantly, in comparison with neurons overexpressing wild-type CaMKIIα, neurons overexpressing a mutant form of the kinase (CaMKII-I205K), unable to interact with GluN2B, were partially protected against OGD-induced damage. Taken together, our results identify crucial determinants in the C-terminal domain of GluN2B subunits in promoting neuronal death in ischemic conditions. These mechanisms underlie the divergent roles of the GluN2A- and GluN2B-NMDARs in determining neuronal fate in cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , Cell Death , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Hippocampus/metabolism , In Vitro Techniques , Mice , Mice, Knockout , Protein Subunits/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics
11.
Neurochem Res ; 41(1-2): 156-82, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26547197

ABSTRACT

Long-term synaptic plasticity in the hippocampus is thought to underlie the formation of certain forms of memory, including spatial memory. The early phase of long-term synaptic potentiation and synaptic depression depends on post-translational modifications of synaptic proteins, while protein synthesis is also required for the late-phase of both forms of synaptic plasticity (L-LTP and L-LTD). Numerous pieces of evidence show a role for different types of proteases in synaptic plasticity, further increasing the diversity of mechanisms involved in the regulation of the intracellular and extracellular protein content. The cleavage of extracellular proteins is coupled to changes in postsynaptic intracellular mechanisms, and additional alterations in this compartment result from the protease-mediated targeting of intracellular proteins. Both mechanisms contribute to initiate signaling cascades that drive downstream pathways coupled to synaptic plasticity. In this review we summarize the evidence pointing to a role for extracellular and intracellular proteases, with distinct specificities, in synaptic plasticity. Where in the cells the proteases are located, and how they are regulated is also discussed. The combined actions of proteases and translation mechanisms contribute to a tight control of the synaptic proteome relevant for long-term synaptic potentiation and synaptic depression in the hippocampus. Additional studies are required to elucidate the mechanisms whereby these changes in the synaptic proteome are related with plasticity phenomena.


Subject(s)
Hippocampus/physiology , Neuronal Plasticity , Peptide Hydrolases/metabolism , Animals , Hippocampus/enzymology , Humans , Proteolysis
12.
Nitric Oxide ; 49: 80-9, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26196856

ABSTRACT

Quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) is a widely used technique to characterize changes in gene expression in complex cellular and tissue processes, such as cytoprotection or inflammation. The accurate assessment of changes in gene expression depends on the selection of adequate internal reference gene(s). Carbon monoxide (CO) affects several metabolic pathways and de novo protein synthesis is crucial in the cellular responses to this gasotransmitter. Herein a selection of commonly used reference genes was analyzed to identify the most suitable internal control genes to evaluate the effect of CO on gene expression in cultured cerebrocortical astrocytes. The cells were exposed to CO by treatment with CORM-A1 (CO releasing molecule A1) and four different algorithms (geNorm, NormFinder, Delta Ct and BestKeeper) were applied to evaluate the stability of eight putative reference genes. Our results indicate that Gapdh (glyceraldehyde-3-phosphate dehydrogenase) together with Ppia (peptidylpropyl isomerase A) is the most suitable gene pair for normalization of qRT-PCR results under the experimental conditions used. Pgk1 (phosphoglycerate kinase 1), Hprt1 (hypoxanthine guanine phosphoribosyl transferase I), Sdha (Succinate Dehydrogenase Complex, Subunit A), Tbp (TATA box binding protein), Actg1 (actin gamma 1) and Rn18s (18S rRNA) genes presented less stable expression profiles in cultured cortical astrocytes exposed to CORM-A1 for up to 60 min. For validation, we analyzed the effect of CO on the expression of Bdnf and bcl-2. Different results were obtained, depending on the reference genes used. A significant increase in the expression of both genes was found when the results were normalized with Gapdh and Ppia, in contrast with the results obtained when the other genes were used as reference. These findings highlight the need for a proper and accurate selection of the reference genes used in the quantification of qRT-PCR results in studies on the effect of CO in gene expression.


Subject(s)
Astrocytes/drug effects , Carbon Monoxide/pharmacology , Cerebral Cortex/cytology , Gene Expression/drug effects , Real-Time Polymerase Chain Reaction/standards , Algorithms , Analysis of Variance , Animals , Astrocytes/metabolism , Boranes/pharmacology , Carbonates/pharmacology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Mice , Rats , Reference Standards , Reproducibility of Results
13.
Biochim Biophys Acta ; 1832(1): 263-74, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23069389

ABSTRACT

Overactivation of glutamate receptors contributes to neuronal damage (excitotoxicity) in ischemic stroke but the detailed mechanisms are not fully elucidated. Brain ischemia is also characterized by an impairment of the activity of the proteasome, one of the major proteolytic systems in neurons. We found that excitotoxic stimulation with glutamate rapidly decreases ATP levels and the proteasome activity, and induces the disassembly of the 26S proteasome in cultured rat hippocampal neurons. Downregulation of the proteasome activity, leading to an accumulation of ubiquitinated proteins, was mediated by calcium entry through NMDA receptors and was only observed in the nuclear fraction. Furthermore, excitotoxicity-induced proteasome inhibition was partially sensitive to cathepsin-L inhibition and was specifically induced by activation of extrasynaptic NMDA receptors. Oxygen and glucose deprivation induced neuronal death and downregulated the activity of the proteasome by a mechanism dependent on the activation of NMDA receptors. Since deubiquitinating enzymes may regulate proteins half-life by counteracting ubiquitination, we also analyzed how their activity is regulated under excitotoxic conditions. Glutamate stimulation decreased the total deubiquitinase activity in hippocampal neurons, but was without effect on the activity of Uch-L1, showing that not all deubiquitinases are affected. These results indicate that excitotoxic stimulation with glutamate has multiple effects on the ubiquitin-proteasome system which may contribute to the demise process in brain ischemia and in other neurological disorders.


Subject(s)
Down-Regulation , Glutamic Acid/metabolism , Hippocampus/metabolism , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Ubiquitin/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain Ischemia/enzymology , Brain Ischemia/genetics , Brain Ischemia/metabolism , Cells, Cultured , Female , Glutamic Acid/toxicity , Hippocampus/cytology , Hippocampus/enzymology , Humans , Male , Neurons/enzymology , Proteasome Endopeptidase Complex/genetics , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/genetics
14.
Neurobiol Dis ; 65: 220-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24513087

ABSTRACT

Cerebral ischemia is characterized by an early disruption of GABAergic neurotransmission contributing to an imbalance of the excitatory/inhibitory equilibrium and neuronal death, but the molecular mechanisms involved are not fully understood. Here we report a downregulation of GABA(A) receptor (GABA(A)R) expression, affecting both mRNA and protein levels of GABA(A)R subunits, in hippocampal neurons subjected to oxygen-glucose deprivation (OGD), an in vitro model of ischemia. Similar alterations in the abundance of GABA(A)R subunits were observed in in vivo brain ischemia. OGD reduced the interaction of surface GABA(A)R with the scaffold protein gephyrin, followed by clathrin-dependent receptor internalization. Internalization of GABA(A)R was dependent on glutamate receptor activation and mediated by dephosphorylation of the ß3 subunit at serine 408/409. Expression of phospho-mimetic mutant GABA(A)R ß3 subunits prevented receptor internalization and protected hippocampal neurons from ischemic cell death. The results show a key role for ß3 GABA(A)R subunit dephosphorylation in the downregulation of GABAergic synaptic transmission in brain ischemia, contributing to neuronal death. GABA(A)R phosphorylation might be a therapeutic target to preserve synaptic inhibition in brain ischemia.


Subject(s)
Glucose/deficiency , Hypoxia/pathology , Infarction, Middle Cerebral Artery/pathology , Neurons/metabolism , Neurons/pathology , Receptors, GABA-B/metabolism , Animals , Calpain/pharmacology , Cell Death/physiology , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Disease Models, Animal , Embryo, Mammalian , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hippocampus/cytology , Humans , Phosphorylation/drug effects , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Rats, Wistar , Receptors, GABA-B/genetics , Time Factors
15.
J Neurosci ; 32(13): 4610-22, 2012 Mar 28.
Article in English | MEDLINE | ID: mdl-22457507

ABSTRACT

Brain-derived neurotrophic factor (BDNF) plays an important role in neuronal survival through activation of TrkB receptors. The trkB gene encodes a full-length receptor tyrosine kinase (TrkB.FL) and its truncated (T1/T2) isoforms. We investigated the changes in TrkB protein levels and signaling activity under excitotoxic conditions, which are characteristic of brain ischemia, traumatic brain injury, and neurodegenerative disorders. Excitotoxic stimulation of cultured rat hippocampal or striatal neurons downregulated TrkB.FL and upregulated a truncated form of the receptor (TrkB.T). Downregulation of TrkB.FL was mediated by calpains, whereas the increase in TrkB.T protein levels required transcription and translation activities. Downregulation of TrkB.FL receptors in hippocampal neurons correlated with a decrease in BDNF-induced activation of the Ras/ERK and PLCγ pathways. However, calpain inhibition, which prevents TrkB.FL degradation, did not preclude the decrease in signaling activity of these receptors. On the other hand, incubation with anisomycin, to prevent the upregulation of TrkB.T, protected to a large extent the TrkB.FL signaling activity, suggesting that truncated receptors may act as dominant-negatives. The upregulation of TrkB.T under excitotoxic conditions was correlated with an increase in BDNF-induced inhibition of RhoA, a mediator of excitotoxic neuronal death. BDNF fully protected hippocampal neurons transduced with TrkB.T when present during excitotoxic stimulation with glutamate, in contrast with the partial protection observed in cells overexpressing TrkB.FL or expressing GFP. These results indicate that BDNF protects hippocampal neurons by two distinct mechanisms: through the neurotrophic effects of TrkB.FL receptors and by activation of TrkB.T receptors coupled to inhibition of the excitotoxic signaling.


Subject(s)
Cell Death/drug effects , Corpus Striatum/metabolism , Gene Expression Regulation/drug effects , Hippocampus/metabolism , Kainic Acid/toxicity , Receptor, trkB/metabolism , Animals , Anisomycin/pharmacology , Brain-Derived Neurotrophic Factor/physiology , Calpain/antagonists & inhibitors , Calpain/physiology , Cell Death/genetics , Corpus Striatum/drug effects , Embryo, Mammalian , Glutamic Acid/toxicity , Hippocampus/drug effects , Isoenzymes/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Primary Cell Culture , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/physiology
16.
J Biol Chem ; 287(9): 6868-77, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22223644

ABSTRACT

Glutamate receptors of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) type mediate fast excitatory synaptic transmission in the CNS. Synaptic strength is modulated by AMPA receptor binding partners, which regulate receptor synaptic targeting and functional properties. We identify Contactin-associated protein 1 (Caspr1) as an AMPA receptor interactor. Caspr1 is present in synapses and interacts with AMPA receptors in brain synaptic fractions. Coexpression of Caspr1 with GluA1 increases the amplitude of glutamate-evoked currents. Caspr1 overexpression in hippocampal neurons increases the number and size of synaptic GluA1 clusters, whereas knockdown of Caspr1 decreases the intensity of synaptic GluA1 clusters. Hence, Caspr1 is a regulator of the trafficking of AMPA receptors to synapses.


Subject(s)
Brain/cytology , Cell Adhesion Molecules, Neuronal/metabolism , Neuronal Plasticity/physiology , Receptors, AMPA/metabolism , Synapses/metabolism , Animals , Brain/metabolism , COS Cells , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Chlorocebus aethiops , Dendrites/metabolism , Gene Knockdown Techniques , Glutamic Acid/pharmacology , HEK293 Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Neurons/metabolism , Neurons/ultrastructure , Protein Transport/physiology , Rats , Rats, Wistar , Receptors, AMPA/genetics , Synaptosomes/metabolism
17.
Cell Mol Life Sci ; 69(16): 2691-715, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22382927

ABSTRACT

In addition to its central roles in protein quality control, regulation of cell cycle, intracellular signaling, DNA damage response and transcription regulation, the ubiquitin-proteasome system (UPS) plays specific roles in the nervous system, where it contributes to precise connectivity through development, and later assures functionality by regulating a wide spectrum of neuron-specific cellular processes. Aberrations in this system have been implicated in the etiology of neurodevelopmental and neurodegenerative diseases. In this review, we provide an updated view on the UPS and highlight recent findings concerning its role in normal and diseased nervous systems. We discuss the advantages of the model organism Caenorhabditis elegans as a tool to unravel the major unsolved questions concerning this biochemical pathway and its involvement in nervous system function and dysfunction, and expose the new possibilities, using state-of-the-art techniques, to assess UPS function using this model system.


Subject(s)
Caenorhabditis elegans/metabolism , Nervous System Physiological Phenomena , Neurodegenerative Diseases/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , Caenorhabditis elegans/genetics
18.
Mol Neurobiol ; 60(6): 3507-3521, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36882590

ABSTRACT

Learned fear is orchestrated by a brain fear network that comprises the amygdala, hippocampus and the medial prefrontal cortex. Synaptic plasticity within this network is critical for the formation of proper fear memories. Known for their role in the promotion of synaptic plasticity, neurotrophins position as obvious candidates in the regulation of fear processes. Indeed, recent evidence from our laboratory and others associates dysregulated signalling through neurotrophin-3 and its receptor TrkC with the pathophysiology of anxiety and fear-related disorders. Here, we put wild-type C57Bl/6J mice through a contextual fear conditioning paradigm in order to characterize TrkC activation and expression in the main brain regions involved in (learned) fear - amygdala, hippocampus, and prefrontal cortex - during the formation of a fear memory. We report an overall decreased activation of TrkC in the fear network during fear consolidation and reconsolidation. During reconsolidation, hippocampal TrkC downregulation was accompanied by a decrease in the expression and activation of Erk, a critical signalling pathway in fear conditioning. Moreover, we did not find evidence that the observed decrease of TrkC activation was caused by altered expression of dominant negative form of TrkC, neurotrophin-3, or the PTP1B phosphatase. Our results indicate hippocampal TrkC inactivation through Erk signalling as a potential mechanism in the regulation of contextual fear memory formation.


Subject(s)
Brain , Fear , Animals , Mice , Brain/metabolism , Fear/physiology , Hippocampus/metabolism , Nerve Growth Factors/metabolism , Prefrontal Cortex/metabolism , Receptor, trkC/metabolism
19.
World J Pediatr ; 19(6): 505-548, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37084165

ABSTRACT

BACKGROUND: Current diagnostic criteria for hypoxic-ischemic encephalopathy in the early hours lack objective measurement tools. Therefore, this systematic review aims to identify putative molecules that can be used in diagnosis in daily clinical practice (PROSPERO ID: CRD42021272610). DATA SOURCES: Searches were performed in PubMed, Web of Science, and Science Direct databases until November 2020. English original papers analyzing samples from newborns > 36 weeks that met at least two American College of Obstetricians and Gynecologists diagnostic criteria and/or imaging evidence of cerebral damage were included. Bias was assessed by the Newcastle-Ottawa Scale. The search and data extraction were verified by two authors separately. RESULTS: From 373 papers, 30 met the inclusion criteria. Data from samples collected in the first 72 hours were extracted, and increased serum levels of neuron-specific enolase and S100-calcium-binding protein-B were associated with a worse prognosis in newborns that suffered an episode of perinatal asphyxia. In addition, the levels of glial fibrillary acidic protein, ubiquitin carboxyl terminal hydrolase isozyme-L1, glutamic pyruvic transaminase-2, lactate, and glucose were elevated in newborns diagnosed with hypoxic-ischemic encephalopathy. Moreover, pathway analysis revealed insulin-like growth factor signaling and alanine, aspartate and glutamate metabolism to be involved in the early molecular response to insult. CONCLUSIONS: Neuron-specific enolase and S100-calcium-binding protein-B are potential biomarkers, since they are correlated with an unfavorable outcome of hypoxic-ischemic encephalopathy newborns. However, more studies are required to determine the sensitivity and specificity of this approach to be validated for clinical practice.


Subject(s)
Asphyxia Neonatorum , Hypoxia-Ischemia, Brain , Pregnancy , Female , Humans , Infant, Newborn , Hypoxia-Ischemia, Brain/diagnosis , Biomarkers , Prognosis , Asphyxia Neonatorum/complications , Asphyxia Neonatorum/diagnosis , S100 Proteins , Phosphopyruvate Hydratase
20.
J Neurosci ; 31(12): 4622-35, 2011 Mar 23.
Article in English | MEDLINE | ID: mdl-21430162

ABSTRACT

GABA is the major inhibitory neurotransmitter in the CNS and changes in GABAergic neurotransmission affect the overall activity of neuronal networks. The uptake of GABA into synaptic vesicles is mediated by the vesicular GABA transporter (VGAT), and changes in the expression of the transporter directly regulate neurotransmitter release. In this work we investigated the changes in VGAT protein levels during ischemia and in excitotoxic conditions, which may affect the demise process. We found that VGAT is cleaved by calpains following excitotoxic stimulation of hippocampal neurons with glutamate, giving rise to a stable truncated cleavage product (tVGAT). VGAT cleavage was also observed after transient middle cerebral artery occlusion in mice, a cerebral ischemia model, and following intrahippocampal injection of kainate, but no effect was observed in transgenic mice overexpressing calpastatin, a calpain inhibitor. Incubation of isolated cerebrocortical synaptic vesicles with recombinant calpain also induced the cleavage of VGAT and formation of stable tVGAT. Immunoblot experiments using antibodies targeting different regions of VGAT and N-terminal sequencing analysis showed that calpain cleaves the transporter in the N-terminal region, at amino acids 52 and 60. Immunocytochemistry of GABAergic striatal neurons expressing GFP fusion proteins with the full-length VGAT or tVGAT showed that cleavage of the transporter induces a loss of synaptic delivery, leading to a homogeneous distribution of the protein along neurites. Our results show that excitotoxicity downregulates full-length VGAT, with a concomitant generation of tVGAT, which is likely to affect GABAergic neurotransmission and may influence cell death during ischemia.


Subject(s)
Neurotoxins/pharmacology , Synapses/metabolism , Vesicular Glutamate Transport Proteins/metabolism , Animals , Blotting, Western , Brain Ischemia/metabolism , Brain Ischemia/pathology , Calpain/metabolism , DNA/genetics , Excitatory Amino Acid Agonists/pharmacology , Female , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Kainic Acid/pharmacology , Long-Term Potentiation/drug effects , Mice , Mice, Inbred C57BL , PC12 Cells , Phosphoric Monoester Hydrolases/metabolism , Plasmids/genetics , Pregnancy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Status Epilepticus/metabolism , Status Epilepticus/pathology , Synapses/drug effects , Synaptic Transmission/physiology , Transfection , gamma-Aminobutyric Acid/physiology
SELECTION OF CITATIONS
SEARCH DETAIL