Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Radiol Case Rep ; 18(3): 932-935, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36593917

ABSTRACT

We present the clinical case of a 53-year-old woman referred for suspicion of recurrence of a mesonephric-like adenocarcinoma of the ovary. Abdominal and pelvic CT revealed multiple round/oval solid nodules with similar density scattered throughout the abdomen and pelvis, the biggest ones appearing in the left hypochondrium; no normal-appearing spleen or ascites were observed. These radiological findings and the absence of significant elevation of CA 125 levels made the radiologists hypothesize that these aspects were related to abdominal splenosis. They asked the patient about previous medical history of splenic injury, which she confirmed, referring it was a consequence of a remote major trauma. A 99mTc-labeled heat-denatured erythrocytes (99mTc-DRBC) scintigraphy/ hybrid SPECT/CT was then performed for definitive diagnosis; it showed spleen remnants as foci of increased radiopharmaceutical uptake in the same locations as the nodules appearing in the CT. This diagnostic work-up was consistent with abdominal splenosis, mimicking peritoneal carcinomatosis of ovarian cancer.

2.
Purinergic Signal ; 16(4): 503-518, 2020 12.
Article in English | MEDLINE | ID: mdl-33025424

ABSTRACT

Caffeine, a stimulant largely consumed around the world, is a non-selective adenosine receptor antagonist, and therefore caffeine actions at synapses usually, but not always, mirror those of adenosine. Importantly, different adenosine receptors with opposing regulatory actions co-exist at synapses. Through both inhibitory and excitatory high-affinity receptors (A1R and A2R, respectively), adenosine affects NMDA receptor (NMDAR) function at the hippocampus, but surprisingly, there is a lack of knowledge on the effects of caffeine upon this ionotropic glutamatergic receptor deeply involved in both positive (plasticity) and negative (excitotoxicity) synaptic actions. We thus aimed to elucidate the effects of caffeine upon NMDAR-mediated excitatory post-synaptic currents (NMDAR-EPSCs), and its implications upon neuronal Ca2+ homeostasis. We found that caffeine (30-200 µM) facilitates NMDAR-EPSCs on pyramidal CA1 neurons from Balbc/ByJ male mice, an action mimicked, as well as occluded, by 1,3-dipropyl-cyclopentylxantine (DPCPX, 50 nM), thus likely mediated by blockade of inhibitory A1Rs. This action of caffeine cannot be attributed to a pre-synaptic facilitation of transmission because caffeine even increased paired-pulse facilitation of NMDA-EPSCs, indicative of an inhibition of neurotransmitter release. Adenosine A2ARs are involved in this likely pre-synaptic action since the effect of caffeine was mimicked by the A2AR antagonist, SCH58261 (50 nM). Furthermore, caffeine increased the frequency of Ca2+ transients in neuronal cell culture, an action mimicked by the A1R antagonist, DPCPX, and prevented by NMDAR blockade with AP5 (50 µM). Altogether, these results show for the first time an influence of caffeine on NMDA receptor activity at the hippocampus, with impact in neuronal Ca2+ homeostasis.


Subject(s)
Caffeine/pharmacology , Hippocampus/drug effects , Neurons/drug effects , Purinergic P1 Receptor Antagonists/pharmacology , Synaptic Transmission/drug effects , Animals , Excitatory Postsynaptic Potentials/drug effects , Glutamine , Hippocampus/metabolism , Male , Mice , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism
3.
J Caffeine Adenosine Res ; 10(2): 45-60, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32566903

ABSTRACT

Adenosine is an endogenous anticonvulsant and neuroprotectant of the brain. Seizure activity produces large quantities of adenosine, and it is this seizure-induced adenosine surge that normally stops a seizure. However, within the context of epilepsy, adenosine plays a wide spectrum of different roles. It not only controls seizures (ictogenesis), but also plays a major role in processes that turn a normal brain into an epileptic brain (epileptogenesis). It is involved in the control of abnormal synaptic plasticity and neurodegeneration and plays a major role in the expression of comorbid symptoms and complications of epilepsy, such as sudden unexpected death in epilepsy (SUDEP). Given the important role of adenosine in epilepsy, therapeutic strategies are in development with the goal to utilize adenosine augmentation not only for the suppression of seizures but also for disease modification and epilepsy prevention, as well as strategies to block adenosine A2A receptor overfunction associated with neurodegeneration. This review provides a comprehensive overview of the role of adenosine in epilepsy.

4.
Br J Pharmacol ; 175(23): 4386-4397, 2018 12.
Article in English | MEDLINE | ID: mdl-30220081

ABSTRACT

BACKGROUND AND PURPOSE: NMDA receptors play a key role in both synaptic plasticity and neurodegeneration. Adenosine is an endogenous neuromodulator and through membrane receptors of the A2A subtype can influence both synaptic plasticity and neuronal death. The present work was designed to evaluate the influence of adenosine A2A receptors upon NMDA receptor activity in CA1 hippocampal neurons. We discriminated between modulation of synaptic versus extrasynaptic receptors, since extrasynaptic NMDA receptors are mostly associated with neurodegeneration while synaptic NMDA receptors are linked to plasticity phenomena. EXPERIMENTAL APPROACH: Whole-cell patch-clamp recordings were obtained to evaluate NMDA receptor actions on CA1 pyramidal neurons of young adult (5-10 weeks) male Wistar rat hippocampus. KEY RESULTS: Activation of A2A receptors with CGS 21680 (30 nM) consistently facilitated chemically-evoked NMDA receptor-currents (NMDA-PSCs) and afferent-evoked NMDA-currents (NMDA-EPSCs), an action prevented by an A2A receptor antagonist (SCH58261, 100 nM) and a PKA inhibitor, H-89 (1 µM). These actions did not reflect facilitation in glutamate release since there was no change in NMDA-EPSCs paired pulse ratio. A2A receptor actions were lost in the presence of an open-channel NMDA receptor blocker, MK-801 (10 µM), but persisted in the presence of memantine, at a concentration (10 µM) known to preferentially block extrasynaptic NMDA receptors. CONCLUSION AND IMPLICATIONS: These results show that A2A receptors exert a positive postsynaptic modulatory effect over synaptic, but not extrasynaptic, NMDA receptors in CA1 neurons and, therefore, under non-pathological conditions may contribute to shift the dual role of NMDA receptors towards enhancement of synaptic plasticity.


Subject(s)
CA1 Region, Hippocampal/metabolism , N-Methylaspartate/metabolism , Pyramidal Cells/metabolism , Receptor, Adenosine A2A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Male , Rats , Rats, Wistar
5.
J Neuroinflammation ; 15(1): 203, 2018 Jul 11.
Article in English | MEDLINE | ID: mdl-29996878

ABSTRACT

BACKGROUND: Epilepsy is a prevalent neurological disorder worldwide. It is characterized by an enduring predisposition to generate seizures and its development is accompanied by alterations in many cellular processes. Organotypic slice cultures represent a multicellular environment with the potential to assess biological mechanisms, and they are used as a starting point for refining molecules for in vivo studies. Here, we investigated organotypic slice cultures as a model of epilepsy. METHODS: We assessed, by electrophysiological recordings, the spontaneous activity of organotypic slices maintained under different culture protocols. Moreover, we evaluated, through molecular-based approaches, neurogenesis, neuronal death, gliosis, expression of proinflammatory cytokines, and activation of NLRP3 inflammasome (nucleotide-binding, leucine-rich repeat, pyrin domain) as biomarkers of neuroinflammation. RESULTS: We demonstrated that organotypic slices, maintained under a serum deprivation culture protocol, develop epileptic-like activity. Furthermore, throughout a comparative study with slices that do not depict any epileptiform activity, slices with epileptiform activity were found to display significant differences in terms of inflammation-related features, such as (1) increased neuronal death, with higher incidence in CA1 pyramidal neurons of the hippocampus; (2) activation of astrocytes and microglia, assessed through western blot and immunohistochemistry; (3) upregulation of proinflammatory cytokines, specifically interleukin-1ß (IL-1ß), interleukin-6, and tumor necrosis factor α, revealed by qPCR; and (4) enhanced expression of NLRP3, assessed by western blot, together with increased NLRP3 activation, showed by IL-1ß quantification. CONCLUSIONS: Thus, organotypic slice cultures gradually deprived of serum mimic the epileptic-like activity, as well as the inflammatory events associated with in vivo epilepsy. This system can be considered a new tool to explore the interplay between neuroinflammation and epilepsy and to screen potential drug candidates, within the inflammatory cascades, to reduce/halt epileptogenesis.


Subject(s)
Anticonvulsants/therapeutic use , Cytokines/metabolism , Epilepsy/drug therapy , Epilepsy/pathology , Hippocampus/drug effects , Action Potentials/drug effects , Animals , Animals, Newborn , Boron Compounds/metabolism , Calcium-Binding Proteins/metabolism , Caspase 3/metabolism , Culture Media, Serum-Free/toxicity , Cytokines/genetics , Disease Models, Animal , Doublecortin Domain Proteins , Epilepsy/chemically induced , Epilepsy/complications , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/etiology , Gliosis/pathology , Hippocampus/pathology , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Organ Culture Techniques , Pregnancy , Rats , Rats, Sprague-Dawley , Spectrin/metabolism
6.
Cereb Cortex ; 28(8): 2795-2809, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29053799

ABSTRACT

The cytokine erythropoietin (EPO) is the master regulator of erythropoiesis. Intriguingly, many studies have shown that the cognitive performance of patients receiving EPO for its hematopoietic effects is enhanced, which prompted the growing interest in the use of EPO-based strategies to treat neuropsychiatric disorders. EPO plays key roles in brain development and maturation, but also modulates synaptic transmission. However, the mechanisms underlying the latter have remained elusive. Here, we show that acute (40-60 min) exposure to EPO presynaptically downregulates spontaneous and afferent-evoked excitatory transmission, without affecting basal firing of action potentials. Conversely, prolonged (3 h) exposure to EPO, if followed by a recovery period (1 h), is able to elicit a homeostatic increase in excitatory spontaneous, but not in evoked, synaptic transmission. These data lend support to the emerging view that segregated pathways underlie spontaneous and evoked neurotransmitter release. Furthermore, we show that prolonged exposure to EPO facilitates a form of hippocampal long-term potentiation that requires noncanonical recruitment of calcium-permeable AMPA receptors for its maintenance. These findings provide important new insight into the mechanisms by which EPO enhances neuronal function, learning, and memory.


Subject(s)
Erythropoietin/pharmacology , Hippocampus/cytology , Hippocampus/physiology , Homeostasis/drug effects , Neuronal Plasticity/drug effects , Synapses/drug effects , Animals , Biophysics , Electric Stimulation , In Vitro Techniques , Long-Term Potentiation/drug effects , Membrane Potentials/drug effects , Nerve Net/drug effects , Nerve Net/physiology , Neurotransmitter Agents/pharmacology , Organ Culture Techniques , Patch-Clamp Techniques , Rats , Receptors, AMPA/metabolism , Receptors, Erythropoietin/metabolism , Sodium Channel Blockers/pharmacology , Synapses/physiology , Tetrodotoxin/pharmacology , Time Factors
7.
J Neurochem ; 139(6): 1056-1070, 2016 12.
Article in English | MEDLINE | ID: mdl-27778347

ABSTRACT

Physiological network functioning in the hippocampus is dependent on a balance between glutamatergic cell excitability and the activity of diverse local circuit neurons that release the inhibitory neurotransmitter γ-aminobutyric acid (GABA). Tuners of neuronal communication such as adenosine, an endogenous modulator of synapses, control hippocampal network operations by regulating excitability. Evidence has been recently accumulating on the influence of adenosine on different aspects of GABAergic transmission to shape hippocampal function. This review addresses how adenosine, through its high-affinity A1 (A1 R) and A2A receptors (A2A R), interferes with different GABA-mediated forms of inhibition in the hippocampus to regulate neuronal excitability. Adenosine-mediated modulation of phasic/tonic inhibitory transmission, of GABA transport mechanisms and its interference with other modulatory systems are discussed together with the putative implications for neuronal function in physiological and pathological conditions. This article is part of a mini review series: 'Synaptic Function and Dysfunction in Brain Diseases'.


Subject(s)
Adenosine/metabolism , GABAergic Neurons/metabolism , Hippocampus/metabolism , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Adenosine/pharmacology , Animals , Hippocampus/drug effects , Humans , Nerve Net/drug effects , Nerve Net/metabolism , Receptors, Purinergic P1/metabolism , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/pharmacology
8.
Purinergic Signal ; 12(2): 283-94, 2016 06.
Article in English | MEDLINE | ID: mdl-26897393

ABSTRACT

Brain-derived neurotrophic factor (BDNF) and adenosine are widely recognized as neuromodulators of glutamatergic transmission in the adult brain. Most BDNF actions upon excitatory plasticity phenomena are under control of adenosine A2A receptors (A2ARs). Concerning gamma-aminobutyric acid (GABA)-mediated transmission, the available information refers to the control of GABA transporters. We now focused on the influence of BDNF and the interplay with adenosine on phasic GABAergic transmission. To assess this, we evaluated evoked and spontaneous synaptic currents recorded from CA1 pyramidal cells in acute hippocampal slices from adult rat brains (6 to 10 weeks old). BDNF (10-100 ng/mL) increased miniature inhibitory postsynaptic current (mIPSC) frequency, but not amplitude, as well as increased the amplitude of inhibitory postsynaptic currents (IPSCs) evoked by afferent stimulation. The facilitatory action of BDNF upon GABAergic transmission was lost in the presence of a Trk inhibitor (K252a, 200 nM), but not upon p75(NTR) blockade (anti-p75(NTR) IgG, 50 µg/mL). Moreover, the facilitatory action of BDNF onto GABAergic transmission was also prevented upon A2AR antagonism (SCH 58261, 50 nM). We conclude that BDNF facilitates GABAergic signaling at the adult hippocampus via a presynaptic mechanism that depends on TrkB and adenosine A2AR activation.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , CA1 Region, Hippocampal/metabolism , Receptor, Adenosine A2A/metabolism , Receptor, trkB/metabolism , Synaptic Transmission/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Inhibitory Postsynaptic Potentials/physiology , Male , Organ Culture Techniques , Patch-Clamp Techniques , Rats , Rats, Wistar
9.
Cereb Cortex ; 26(3): 1081-95, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25452570

ABSTRACT

Adenosine is an endogenous neuromodulator that decreases excitability of hippocampal circuits activating membrane-bound metabotropic A1 receptor (A1R). The presynaptic inhibitory action of adenosine A1R in glutamatergic synapses is well documented, but its influence on inhibitory GABAergic transmission is poorly known. We report that GABAA receptor (GABAAR)-mediated tonic, but not phasic, transmission is suppressed by A1R in hippocampal neurons. Adenosine A1R activation strongly inhibits GABAAR agonist (muscimol)-evoked currents in Cornu Ammonis 1 (CA1) pyramidal neurons and in a specific subpopulation of interneurons expressing axonal cannabinoid receptor type 1. In addition, A1R suppresses tonic GABAAR currents measured in the presence of elevated ambient GABA as well as in naïve slices. The inhibition of GABAergic currents involves both protein kinase A (PKA) and protein kinase C (PKC) signaling pathways and decreases GABAAR δ-subunit expression. On the contrary, no A1R-mediated modulation was detected in phasic inhibitory postsynaptic currents evoked either by afferent electrical stimulation or by spontaneous quantal release. The results show that A1R modulates extrasynaptic rather than synaptic GABAAR-mediated signaling, and that this modulation selectively occurs in hippocampal pyramidal neurons and in a specific subpopulation of inhibitory interneurons. We conclude that modulation of tonic GABAAR signaling by adenosine A1R in specific neuron types may regulate neuronal gain and excitability in the hippocampus.


Subject(s)
CA1 Region, Hippocampal/physiology , Interneurons/physiology , Pyramidal Cells/physiology , Receptor, Adenosine A1/metabolism , Receptors, GABA-A/metabolism , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Immunoblotting , Immunohistochemistry , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Interneurons/cytology , Interneurons/drug effects , Male , Miniature Postsynaptic Potentials/drug effects , Miniature Postsynaptic Potentials/physiology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Patch-Clamp Techniques , Protein Kinase C/metabolism , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Rats, Wistar , Tissue Culture Techniques
10.
Biotechnol J ; 10(10): 1578-88, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26123315

ABSTRACT

Standardization of culture methods for human pluripotent stem cell (PSC) neural differentiation can greatly contribute to the development of novel clinical advancements through the comprehension of neurodevelopmental diseases. Here, we report an approach that reproduces neural commitment from human induced pluripotent stem cells using dual-SMAD inhibition under defined conditions in a vitronectin-based monolayer system. By employing this method it was possible to obtain neurons derived from both control and Rett syndrome patients' pluripotent cells. During differentiation mutated cells displayed alterations in the number of neuronal projections, and production of Tuj1 and MAP2-positive neurons. Although investigation of a broader number of patients would be required, these observations are in accordance with previous studies showing impaired differentiation of these cells. Consequently, our experimental methodology was proved useful not only for the generation of neural cells, but also made possible to compare neural differentiation behavior of different cell lines under defined culture conditions. This study thus expects to contribute with an optimized approach to study the neural commitment of human PSCs, and to produce patient-specific neural cells that can be used to gain a better understanding of disease mechanisms.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/genetics , Induced Pluripotent Stem Cells/cytology , Neurogenesis , Rett Syndrome/genetics , Cell Line , Cell Proliferation/genetics , Culture Media , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Humans , Methyl-CpG-Binding Protein 2/biosynthesis , Methyl-CpG-Binding Protein 2/genetics , Neural Stem Cells/cytology , Neurons/cytology , Rett Syndrome/pathology , Rett Syndrome/therapy , Smad Proteins, Inhibitory/genetics
11.
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
12.
Hippocampus ; 25(5): 566-80, 2015 May.
Article in English | MEDLINE | ID: mdl-25402014

ABSTRACT

Adenosine inhibits excitatory neurons widely in the brain through adenosine A1 receptor, but activation of adenosine A2A receptor (A2A R) has an opposite effect promoting discharge in neuronal networks. In the hippocampus A2A R expression level is low, and the receptor's effect on identified neuronal circuits is unknown. Using optogenetic afferent stimulation and whole-cell recording from identified postsynaptic neurons we show that A2A R facilitates excitatory glutamatergic Schaffer collateral synapses to CA1 pyramidal cells, but not to GABAergic inhibitory interneurons. In addition, A2A R enhances GABAergic inhibitory transmission between CA1 area interneurons leading to disinhibition of pyramidal cells. Adenosine A2A R has no direct modulatory effect on GABAergic synapses to pyramidal cells. As a result adenosine A2A R activation alters the synaptic excitation - inhibition balance in the CA1 area resulting in increased pyramidal cell discharge to glutamatergic Schaffer collateral stimulation. In line with this, we show that A2A R promotes synchronous pyramidal cell firing in hyperexcitable conditions where extracellular potassium is elevated or following high-frequency electrical stimulation. Our results revealed selective synapse- and cell type specific adenosine A2A R effects in hippocampal CA1 area. The uncovered mechanisms help our understanding of A2A R's facilitatory effect on cortical network activity.


Subject(s)
CA1 Region, Hippocampal/physiology , Receptor, Adenosine A2A/metabolism , Synapses/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , CA1 Region, Hippocampal/drug effects , Electric Stimulation , Extracellular Space/metabolism , Glutamic Acid/metabolism , Interneurons/drug effects , Interneurons/physiology , Mice, Transgenic , Neural Inhibition/drug effects , Neural Inhibition/physiology , Optogenetics , Patch-Clamp Techniques , Potassium/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Synapses/drug effects , Tissue Culture Techniques , gamma-Aminobutyric Acid/metabolism
13.
Trends Neurosci ; 36(4): 248-57, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23332692

ABSTRACT

It is widely accepted that Hebbian forms of plasticity mediate selective modifications in synaptic strength underlying information encoding in response to experience and circuit formation or refinement throughout development. Several complementary forms of homeostatic plasticity coordinate to keep Hebbian plasticity in check, frequently through the actions of conserved regulatory molecules. Recent evidence suggests that this may be the case for adenosine, which is ubiquitous in the brain and is released by both neurons and glial cells via constitutive and activity-dependent mechanisms. Through A1 and A2A receptor activation, adenosine modulates neuronal homeostasis and tunes the ability of synapses to undergo and/or sustain plasticity. Here, we review how adenosine equilibrates neuronal activity and sets the stage for synaptic plasticity.


Subject(s)
Adenosine/metabolism , Neuronal Plasticity/physiology , Neurons/cytology , Synapses/physiology , Animals , Brain/cytology , Humans , Neurons/physiology , Receptors, Purinergic P1/metabolism
14.
Neuropharmacology ; 65: 114-22, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23041538

ABSTRACT

Long lasting enhancement of synaptic transmission can be triggered by brief bursts of afferent stimulation, underlying long-term potentiation (LTP), and also by brief ischemia in a process known as i-LTP. The extent to which LTP and i-LTP rely on comparable cellular mechanisms remains unclear. Under physiological conditions, LTP induction drives transient expression of calcium-permeable AMPARs (CP-AMPARs) at synapses, whose ability to undergo plasticity is primed by endogenous activation of adenosine A(2A) receptors (A(2A)Rs). The present work thus addressed the contribution of CP-AMPARs and A(2A)Rs to i-LTP, which was induced in rat hippocampal slices by brief (10 min) oxygen/glucose deprivation (OGD). The amplitude of afferent-evoked excitatory postsynaptic currents (EPSCs) recorded from CA1 pyramidal neurons was decreased during OGD but gradually recovered toward values significantly above (157 ± 17%) the baseline (100%) 40-50 min after re-oxygenation. This i-LTP was precluded by CP-AMPAR blockade (internal spermine (500 µM) or extracellular NASPM (20 µM) application) as well as by A(2A)R blockade with a selective antagonist (SCH 58261, 100 nM). OGD prompted sustained (>70 min) facilitation of mEPSC amplitude and frequency, and decreased mEPSC decay time, all of which were prevented by SCH 58261 (100 nM). The ability of NASPM (20 µM) to acutely inhibit EPSCs 1 h after OGD, but not in control conditions nor in OGD-challenged slices when in the presence of SCH 58261 (100 nM), further supports sustained CP-AMPAR recruitment by i-LTP in an A(2A)R-dependent way. We propose that although i-LTP may initially mimic LTP, failure of auto-regulated CP-AMPAR removal from synapses could constitute an early divergent event between these forms of plasticity.


Subject(s)
Calcium/metabolism , Hippocampus/blood supply , Hippocampus/metabolism , Neuronal Plasticity/physiology , Receptors, AMPA/biosynthesis , Synapses/metabolism , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Gene Expression Regulation , Hippocampus/drug effects , Male , Neuronal Plasticity/drug effects , Organ Culture Techniques , Pyrimidines/pharmacology , Rats , Rats, Wistar , Receptors, AMPA/agonists , Receptors, AMPA/antagonists & inhibitors , Spermine/pharmacology , Synapses/drug effects , Triazoles/pharmacology
15.
J Neurosci ; 32(34): 11750-62, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22915117

ABSTRACT

Parkinson's disease (PD) is the most common representative of a group of disorders known as synucleinopathies, in which misfolding and aggregation of α-synuclein (a-syn) in various brain regions is the major pathological hallmark. Indeed, the motor symptoms in PD are caused by a heterogeneous degeneration of brain neurons not only in substantia nigra pars compacta but also in other extrastriatal areas of the brain. In addition to the well known motor dysfunction in PD patients, cognitive deficits and memory impairment are also an important part of the disorder, probably due to disruption of synaptic transmission and plasticity in extrastriatal areas, including the hippocampus. Here, we investigated the impact of a-syn aggregation on AMPA and NMDA receptor-mediated rat hippocampal (CA3-CA1) synaptic transmission and long-term potentiation (LTP), the neurophysiological basis for learning and memory. Our data show that prolonged exposure to a-syn oligomers, but not monomers or fibrils, increases basal synaptic transmission through NMDA receptor activation, triggering enhanced contribution of calcium-permeable AMPA receptors. Slices treated with a-syn oligomers were unable to respond with further potentiation to theta-burst stimulation, leading to impaired LTP. Prior delivery of a low-frequency train reinstated the ability to express LTP, implying that exposure to a-syn oligomers drives the increase of glutamatergic synaptic transmission, preventing further potentiation by physiological stimuli. Our novel findings provide mechanistic insight on how a-syn oligomers may trigger neuronal dysfunction and toxicity in PD and other synucleinopathies.


Subject(s)
Long-Term Potentiation/drug effects , Synapses/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , alpha-Synuclein/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Animals , Biophysics , Biotinylation , Cell Line, Tumor , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Extracellular Fluid/metabolism , Hippocampus/cytology , Humans , Insulin/pharmacology , L-Lactate Dehydrogenase/metabolism , Long-Term Potentiation/physiology , Male , Neuroblastoma/pathology , Organ Culture Techniques , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Valine/analogs & derivatives , Valine/pharmacology , alpha-Synuclein/biosynthesis , alpha-Synuclein/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...