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










Publication year range
1.
Sci Adv ; 8(9): eabj0112, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35245123

ABSTRACT

Microglia interact with neurons to facilitate synapse plasticity; however, signal(s) contributing to microglia activation for synapse elimination in pathology are not fully understood. Here, using in vitro organotypic hippocampal slice cultures and transient middle cerebral artery occlusion (MCAO) in genetically engineered mice in vivo, we report that at 24 hours after ischemia, microglia release brain-derived neurotrophic factor (BDNF) to downregulate glutamatergic and GABAergic synapses within the peri-infarct area. Analysis of the cornu ammonis 1 (CA1) in vitro shows that proBDNF and mBDNF downregulate glutamatergic dendritic spines and gephyrin scaffold stability through p75 neurotrophin receptor (p75NTR) and tropomyosin receptor kinase B (TrkB) receptors, respectively. After MCAO, we report that in the peri-infarct area and in the corresponding contralateral hemisphere, similar neuroplasticity occurs through microglia activation and gephyrin phosphorylation at serine-268 and serine-270 in vivo. Targeted deletion of the Bdnf gene in microglia or GphnS268A/S270A (phospho-null) point mutations protects against ischemic brain damage, neuroinflammation, and synapse downregulation after MCAO.


Subject(s)
Brain Ischemia , Brain-Derived Neurotrophic Factor , Animals , Brain-Derived Neurotrophic Factor/genetics , Infarction , Mice , Microglia , Receptor, trkB , Serine , Synapses
2.
Neurobiol Dis ; 130: 104490, 2019 10.
Article in English | MEDLINE | ID: mdl-31175985

ABSTRACT

Christianson Syndrome is a rare but increasingly diagnosed X-linked intellectual disability disorder that arises from mutations in SLC9A6/NHE6, a pH-regulating transporter that localizes to early and recycling endosomes. We have recently reported that one of the originally identified disease-causing mutations in NHE6 (p.E287-S288del, or ΔES) resulted in a loss of its pH regulatory function. However, the impact of this mutation upon neuronal synapse formation and plasticity is unknown. Here, we investigate the consequences of the ΔES mutant upon mouse hippocampal pyramidal neurons by expressing a fluorescently-labeled ΔES NHE6 construct into primary hippocampal neurons. Neurons expressing the ΔES mutant showed significant reductions in mature dendritic spine density with a concurrent increase in immature filopodia. Furthermore, compared to wild-type (WT), ΔES-containing endosomes are redirected away from early and recycling endosomes toward lysosomes. In parallel, the ΔES mutant reduced the trafficking of glutamatergic AMPA receptors to excitatory synapses and increased their accumulation within lysosomes for potential degradation. Upon long-term potentiation (LTP), neurons expressing ΔES failed to undergo significant structural and functional changes as observed in controls and WT transfectants. Interestingly, synapse density and LTP-induced synaptic remodeling in ΔES-expressing neurons were partially restored by bafilomycin, a vesicular alkalinisation agent, or by leupeptin, an inhibitor of lysosomal proteolytic degradation. Overall, our results demonstrate that the ∆ES mutation attenuates synapse density and structural and functional plasticity in hippocampal neurons. These deficits may be partially due to the mistargeting of AMPA receptors and other cargos to lysosomes, thereby preventing their trafficking during synaptic remodeling. This mechanism may contribute to the cognitive learning deficits observed in patients with Christianson Syndrome and suggests a potential therapeutic strategy for treatment.


Subject(s)
Ataxia/genetics , Epilepsy/genetics , Genetic Diseases, X-Linked/genetics , Hippocampus/metabolism , Hippocampus/pathology , Intellectual Disability/genetics , Microcephaly/genetics , Neuronal Plasticity/genetics , Ocular Motility Disorders/genetics , Sodium-Hydrogen Exchangers/genetics , Animals , Dendritic Spines/metabolism , Dendritic Spines/pathology , Mice , Mutation , Protein Transport/genetics , Receptors, AMPA/metabolism
3.
J Physiol ; 596(17): 4253-4267, 2018 09.
Article in English | MEDLINE | ID: mdl-29928778

ABSTRACT

KEY POINTS: Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is an early-onset neurodegenerative human disease characterized in part by ataxia and Purkinje cell loss in anterior cerebellar lobules. A knock-out mouse model has been developed that recapitulates several features of ARSACS. Using this ARSACS mouse model, we report changes in synaptic input and intrinsic firing in cerebellar Purkinje cells, as well as in their synaptic output in the deep cerebellar nuclei. Changes in firing are observed in anterior lobules that later exhibit Purkinje cell death, but not in posterior lobules that do not. Our results show that both synaptic and intrinsic alterations in Purkinje cell properties likely contribute to disease manifestation in ARSACS; these findings resemble pathophysiological changes reported in several other ataxias. ABSTRACT: Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is an early-onset neurodegenerative disease that includes a pronounced and progressive cerebellar dysfunction. ARSACS is caused by an autosomal recessive loss-of-function mutation in the Sacs gene that encodes the protein sacsin. To better understand the cerebellar pathophysiology in ARSACS, we studied synaptic and firing properties of Purkinje cells from a mouse model of ARSACS, Sacs-/- mice. We found that excitatory synaptic drive was reduced onto Sacs-/- Purkinje cells, and that Purkinje cell firing rate, but not regularity, was reduced at postnatal day (P)40, an age when ataxia symptoms were first reported. Firing rate deficits were limited to anterior lobules that later display Purkinje cell death, and were not observed in posterior lobules where Purkinje cells are not lost. Mild firing deficits were observed as early as P20, prior to the manifestation of motor deficits, suggesting that a critical level of cerebellar dysfunction is required for motor coordination to emerge. Finally, we observed a reduction in Purkinje cell innervation onto target neurons in the deep cerebellar nuclei (DCN) in Sacs-/- mice. Together, these findings suggest that multiple alterations in the cerebellar circuit including Purkinje cell input and output contribute to cerebellar-related disease onset in ARSACS.


Subject(s)
Cerebellar Ataxia/physiopathology , Disease Models, Animal , Heat-Shock Proteins/physiology , Muscle Spasticity/physiopathology , Purkinje Cells/physiology , Spinocerebellar Ataxias/congenital , Synapses/physiology , Animals , Behavior, Animal , Humans , Mice , Mice, Knockout , Mutation , Purkinje Cells/cytology , Spinocerebellar Ataxias/physiopathology
4.
ACS Chem Neurosci ; 9(2): 260-271, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29078046

ABSTRACT

Dendritic polyglycerols (dPG), particularly dendritic polyglycerol sulfates (dPGS), have been intensively studied due to their intrinsic anti-inflammatory activity. As related to brain pathologies involving neuroinflammation, the current study examined if dPG and dPGS can (i) regulate neuroglial activation, and (ii) normalize the morphology and function of excitatory postsynaptic dendritic spines adversely affected by the neurotoxic 42 amino acid amyloid-ß (Aß42) peptide of Alzheimer disease (AD). The exact role of neuroglia, such as microglia and astrocytes, remains controversial especially their positive and negative impact on inflammatory processes in AD. To test dPGS effectiveness in AD models we used primary neuroglia and organotypic hippocampal slice cultures exposed to Aß42 peptide. Overall, our data indicate that dPGS is taken up by both microglia and astrocytes in a concentration- and time-dependent manner. The mechanism of action of dPGS involves binding to Aß42, i.e., a direct interaction between dPGS and Aß42 species interfered with Aß fibril formation and reduced the production of the neuroinflammagen lipocalin-2 (LCN2) mainly in astrocytes. Moreover, dPGS normalized the impairment of neuroglia and prevented the loss of dendritic spines at excitatory synapses in the hippocampus. In summary, dPGS has desirable therapeutic properties that may help reduce amyloid-induced neuroinflammation and neurotoxicity in AD.


Subject(s)
Dendrimers/pharmacology , Dendritic Spines/drug effects , Glycerol/analogs & derivatives , Glycerol/pharmacology , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Synapses/drug effects , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dendritic Spines/metabolism , Dendritic Spines/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Lipocalin-2/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Neuroglia/metabolism , Neuroglia/pathology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Peptide Fragments/administration & dosage , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Surface Plasmon Resonance , Synapses/metabolism , Synapses/pathology , Tissue Culture Techniques
5.
Sci Rep ; 5: 15410, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26510576

ABSTRACT

The amyloid-ß42 (Aß42) peptide is believed to be the main culprit in the pathogenesis of Alzheimer disease (AD), impairing synaptic function and initiating neuronal degeneration. Soluble Aß42 oligomers are highly toxic and contribute to progressive neuronal dysfunction, loss of synaptic spine density, and affect long-term potentiation (LTP). We have characterized a short, L-amino acid Aß-oligomer Interacting Peptide (AIP) that targets a relatively well-defined population of low-n Aß42 oligomers, rather than simply inhibiting the aggregation of Aß monomers into oligomers. Our data show that AIP diminishes the loss of Aß42-induced synaptic spine density and rescues LTP in organotypic hippocampal slice cultures. Notably, the AIP enantiomer (comprised of D-amino acids) attenuated the rough-eye phenotype in a transgenic Aß42 fly model and significantly improved the function of photoreceptors of these flies in electroretinography tests. Overall, our results indicate that specifically "trapping" low-n oligomers provides a novel strategy for toxic Aß42-oligomer recognition and removal.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Oligopeptides/pharmacology , Peptide Fragments/antagonists & inhibitors , Protein Aggregation, Pathological/drug therapy , Synapses/metabolism , Synaptic Transmission/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Peptide Fragments/metabolism , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Rats , Rats, Wistar , Synapses/pathology
6.
Biomacromolecules ; 16(9): 3073-82, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26218295

ABSTRACT

Hyperactivity of microglia and loss of functional circuitry is a common feature of many neurological disorders including those induced or exacerbated by inflammation. Herein, we investigate the response of microglia and changes in hippocampal dendritic postsynaptic spines by dendritic polyglycerol sulfate (dPGS) treatment. Mouse microglia and organotypic hippocampal slices were exposed to dPGS and an inflammogen (lipopolysaccharides). Measurements of intracellular fluorescence and confocal microscopic analyses revealed that dPGS is avidly internalized by microglia but not CA1 pyramidal neurons. Concentration and time-dependent response studies consistently showed no obvious toxicity of dPGS. The adverse effects induced by proinflammogen LPS exposure were reduced and dendritic spine morphology was normalized with the addition of dPGS. This was accompanied by a significant reduction in nitrite and proinflammatory cytokines (TNF-α and IL-6) from hyperactive microglia suggesting normalized circuitry function with dPGS treatment. Collectively, these results suggest that dPGS acts anti-inflammatory, inhibits inflammation-induced degenerative changes in microglia phenotype and rescues dendritic spine morphology.


Subject(s)
CA1 Region, Hippocampal/metabolism , Dendritic Spines/metabolism , Glycerol/pharmacology , Microglia/metabolism , Polymers/pharmacology , Pyramidal Cells/metabolism , Animals , Cell Line , Dendritic Spines/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Mice , Mice, Transgenic , Microglia/pathology , Nervous System Diseases/chemically induced , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Pyramidal Cells/pathology , Tumor Necrosis Factor-alpha/metabolism
7.
Crit Care Med ; 43(9): 1859-69, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26102252

ABSTRACT

OBJECTIVES: There are no current pharmacological therapies for the prevention or treatment of acute respiratory distress syndrome. Early dysregulated inflammation likely plays a role in acute respiratory distress syndrome development and possibly acute respiratory distress syndrome outcomes. p38 mitogen-activated protein kinase is central to the regulation of multiple inflammatory mediators implicated in acute organ dysfunction and is the target for a novel class of cytokine-suppressive anti-inflammatory drugs. In preclinical models, p38 inhibitors reduce lung injury following pancreatitis and burn injury. DESIGN: We conducted a phase IIa, randomized, double-blind, placebo-controlled, parallel-group study to evaluate the safety and tolerability of dilmapimod, a novel p38 mitogen-activated protein kinase inhibitor, in patients at risk for developing acute respiratory distress syndrome admitted with an Injury Severity Score more than 16, excluding head trauma. Enrolled patients received 4- or 24-hour IV dilmapimod infusions at different doses or placebo, daily for 3 days, in four separate cohorts. SETTING: Multicenter randomized clinical trial of large, academic trauma centers. MEASUREMENTS AND MAIN RESULTS: Seventy-seven patients were enrolled. Although adverse events were common in this critically ill population, dilmapimod was well tolerated, with no clinically relevant safety findings. Pharmacokinetic models indicated that the higher dose of 10 mg given as continuous infusion over 24 hours had the most favorable plasma concentration profile. Likewise, measures of soluble inflammatory markers including interleukin-6, C-reactive peptide, interleukin-8, and soluble tumor necrosis factor receptor 1 were most different between this dosing arm and placebo. Although the study was not specifically designed with acute respiratory distress syndrome as an outcome, the number of patients who developed acute respiratory distress syndrome was small (2/77). CONCLUSIONS: The novel p38 mitogen-activated protein kinase inhibitor dilmapimod appears well tolerated and may merit further evaluation for prevention of acute respiratory distress syndrome and other organ injury in larger clinical trials. Furthermore, results of this early-phase trial may aid in design of future studies aimed at prevention of acute respiratory distress syndrome and other organ injury.


Subject(s)
Inflammation Mediators/metabolism , Pyridones/administration & dosage , Pyridones/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Respiratory Distress Syndrome/prevention & control , Wounds and Injuries/drug therapy , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adult , C-Reactive Protein/drug effects , Dose-Response Relationship, Drug , Double-Blind Method , Female , Hospital Mortality , Humans , Interleukin-6/biosynthesis , Interleukin-8/drug effects , Male , Middle Aged , Pyridones/adverse effects , Pyrimidines/adverse effects , Receptors, Tumor Necrosis Factor, Type I/drug effects , Trauma Severity Indices
8.
J Neuroinflammation ; 12: 34, 2015 Feb 22.
Article in English | MEDLINE | ID: mdl-25889069

ABSTRACT

BACKGROUND: Recent studies have revealed that excessive activation of microglia and inflammation-mediated neurotoxicity are implicated in the progression of several neurological disorders. In particular, chronic inflammation in vivo and exposure of cultured brain cells to lipopolysaccharide (LPS) in vitro can adversely change microglial morphology and function. This can have both direct and indirect effects on synaptic structures and functions. The integrity of dendritic spines, the postsynaptic component of excitatory synapses, dictates synaptic efficacy. Interestingly, dysgenesis of dendritic spines has been found in many neurological diseases associated with ω-3 polyunsaturated fatty acid (PUFA) deficiency and cognitive decline. In contrast, supplemented ω-3 PUFAs, such as docosahexaenoic acid (DHA), can partly correct spine defects. Hence, we hypothesize that DHA directly affects synaptic integrity and indirectly through neuron-glia interaction. Strong activation of microglia by LPS is accompanied by marked release of nitric oxide and formation of lipid bodies (LBs), both dynamic biomarkers of inflammation. Here we investigated direct effects of DHA on synaptic integrity and its indirect effects via microglia in the hippocampal CA1 region. METHODS: Microglia (N9) and organotypic hippocampal slice cultures were exposed to the proinflammagen LPS (100 ng/ml) for 24 h. Biochemical and morphological markers of inflammation were investigated in microglia and CA1 regions of hippocampal slices. As biomarkers of hyperactive microglia, mitochondrial function, nitric oxide release and LBs (number, size, LB surface-associated proteins) were assessed. Changes in synaptic transmission of CA1 pyramidal cells were determined following LPS and DHA (25-50 µM) treatments by recording spontaneous AMPA-mediated miniature excitatory postsynaptic currents (mEPSCs). RESULTS: Microglia responded to LPS stimulation with a significant decrease of mitochondrial function, increased nitric oxide production and an increase in the formation of large LBs. LPS treatment led to a significant reduction of dendritic spine densities and an increase in the AMPA-mediated mEPSC inter-event interval (IEI). DHA normalized the LPS-induced abnormalities in both neurons and microglia, as revealed by the restoration of synaptic structures and functions in hippocampal CA1 pyramidal neurons. CONCLUSION: Our findings indicate that DHA can prevent LPS-induced abnormalities (neuroinflammation) by reducing inflammatory biomarkers, thereby normalizing microglia activity and their effect on synaptic function.


Subject(s)
Dendritic Spines/drug effects , Docosahexaenoic Acids/pharmacology , Hippocampus/cytology , Microglia/drug effects , Neurons/cytology , Neurons/drug effects , Action Potentials/drug effects , Animals , Animals, Newborn , Benzimidazoles/metabolism , Carbocyanines/metabolism , Cells, Cultured , Cytochromes c/metabolism , Gene Expression Regulation/drug effects , In Vitro Techniques , Lipid Droplets/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Neurons/physiology , Nitric Oxide/metabolism , Organ Culture Techniques , Perilipin-2 , Polysaccharides/pharmacology
9.
Eur J Neurosci ; 40(5): 2766-76, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24925283

ABSTRACT

CX 546, an allosteric positive modulator of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type ionotropic glutamate receptors (AMPARs), belongs to a drug class called ampakines. These compounds have been shown to enhance long-term potentiation (LTP), a cellular model of learning and memory, and improve animal learning task performance, and have augmented cognition in neurodegenerative patients. However, the chronic effect of CX546 on synaptic structures has not been examined. The structure and integrity of dendritic spines are thought to play a role in learning and memory, and their abnormalities have been implicated in cognitive disorders. In addition, their structural plasticity has been shown to be important for cognitive function, such that dendritic spine remodeling has been proposed as the morphological correlate for LTP. Here, we tested the effect of CX546 on dendritic spine remodeling following long-term treatment. We found that, with prolonged CX546 treatment, organotypic hippocampal slice cultures showed a significant reduction in CA3-CA1 excitatory synapse and spine density. Electrophysiological approaches revealed that the CA3-CA1 circuitry compensates for this synapse loss by increasing synaptic efficacy through enhancement of presynaptic release probability. CX546-treated slices showed prolonged and enhanced potentiation upon LTP induction. Furthermore, structural plasticity, namely spine head enlargement, was also more pronounced after CX546 treatment. Our results suggest a concordance of functional and structural changes that is enhanced with prolonged CX546 exposure. Thus, the improved cognitive ability of patients receiving ampakine treatment may result from the priming of synapses through increases in the structural plasticity and functional reliability of hippocampal synapses.


Subject(s)
Dendritic Spines/drug effects , Dioxanes/pharmacology , Excitatory Amino Acid Agents/pharmacology , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Piperidines/pharmacology , Presynaptic Terminals/drug effects , Animals , Cell Survival/drug effects , Dendritic Spines/physiology , Dioxoles , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Hippocampus/physiology , Long-Term Potentiation/physiology , Mice, Transgenic , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Presynaptic Terminals/physiology , Receptors, AMPA/metabolism , Synapses/drug effects , Synapses/physiology , Synaptic Vesicles/drug effects , Synaptic Vesicles/metabolism , Time Factors , Tissue Culture Techniques
10.
Eur J Neurosci ; 38(11): 3554-66, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24118418

ABSTRACT

Brain trauma can disrupt synaptic connections, and this in turn can prompt axons to sprout and form new connections. If these new axonal connections are aberrant, hyperexcitability can result. It has been shown that ablating tropomyosin-related kinase B (TrkB), a receptor for brain-derived neurotrophic factor (BDNF), can reduce axonal sprouting after hippocampal injury. However, it is unknown whether inhibiting BDNF-mediated axonal sprouting will reduce hyperexcitability. Given this, our purpose here was to determine whether pharmacologically blocking BDNF inhibits hyperexcitability after injury-induced axonal sprouting in the hippocampus. To induce injury, we made Schaffer collateral lesions in organotypic hippocampal slice cultures. As reported by others, we observed a 50% reduction in axonal sprouting in cultures treated with a BDNF blocker (TrkB-Fc) 14 days after injury. Furthermore, lesioned cultures treated with TrkB-Fc were less hyperexcitable than lesioned untreated cultures. Using electrophysiology, we observed a two-fold decrease in the number of CA3 neurons that showed bursting responses after lesion with TrkB-Fc treatment, whereas we found no change in intrinsic neuronal firing properties. Finally, evoked field excitatory postsynaptic potential recordings indicated an increase in network activity within area CA3 after lesion, which was prevented with chronic TrkB-Fc treatment. Taken together, our results demonstrate that blocking BDNF attenuates injury-induced hyperexcitability of hippocampal CA3 neurons. Axonal sprouting has been found in patients with post-traumatic epilepsy. Therefore, our data suggest that blocking the BDNF-TrkB signaling cascade shortly after injury may be a potential therapeutic target for the treatment of post-traumatic epilepsy.


Subject(s)
Brain Injuries/physiopathology , Brain-Derived Neurotrophic Factor/deficiency , CA3 Region, Hippocampal/physiology , Excitatory Postsynaptic Potentials , Animals , Axons/pathology , Brain Injuries/metabolism , CA3 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/pathology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/pathology , Pyramidal Cells/physiopathology
11.
PLoS One ; 8(10): e76299, 2013.
Article in English | MEDLINE | ID: mdl-24098468

ABSTRACT

Epilepsy is a chronic brain disorder involving recurring seizures often precipitated by an earlier neuronal insult. The mechanisms that link the transient neuronal insult to the lasting state of epilepsy are unknown. Here we tested the possible role of DNA methylation in mediating long-term induction of epileptiform activity by transient kainic acid exposure using in vitro and in vivo rodent models. We analyzed changes in the gria2 gene, which encodes for the GluA2 subunit of the ionotropic glutamate, alpha-amino-3-hydroxy-5-methyl-4-isoxazole proprionic acid receptor and is well documented to play a role in epilepsy. We show that kainic acid exposure for two hours to mouse hippocampal slices triggers methylation of a 5' regulatory region of the gria2 gene. Increase in methylation persists one week after removal of the drug, with concurrent suppression of gria2 mRNA expression levels. The degree of kainic acid-induced hypermethylation of gria2 5' region varies between individual slices and correlates with the changes in excitability induced by kainic acid. In a rat in vivo model of post kainic acid-induced epilepsy, we show similar hypermethylation of the 5' region of gria2. Inter-individual variations in gria2 methylation, correlate with the frequency and intensity of seizures among epileptic rats. Luciferase reporter assays support a regulatory role for methylation of gria2 5' region. Inhibition of DNA methylation by RG108 blocked kainic acid-induced hypermethylation of gria2 5' region in hippocampal slice cultures and bursting activity. Our results suggest that DNA methylation of such genes as gria2 mediates persistent epileptiform activity and inter-individual differences in the epileptic response to neuronal insult and that pharmacological agents that block DNA methylation inhibit epileptiform activity raising the prospect of DNA methylation inhibitors in epilepsy therapeutics.


Subject(s)
DNA Methylation , Epilepsy/genetics , Genetic Association Studies , Phenotype , 5' Flanking Region , Animals , Cell Line , CpG Islands , DNA Methylation/drug effects , Epilepsy/chemically induced , Epilepsy/diagnosis , Gene Order , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Kainic Acid/adverse effects , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Neurons/drug effects , Neurons/metabolism , Phthalimides/pharmacology , Promoter Regions, Genetic , Rats , Receptors, AMPA/genetics , Seizures/chemically induced , Seizures/diagnosis , Seizures/genetics , Tryptophan/analogs & derivatives , Tryptophan/pharmacology
12.
Eur J Neurosci ; 35(12): 1908-16, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22708602

ABSTRACT

Most excitatory transmission in the brain is mediated by the AMPA receptor subtype of the ionotropic glutamate receptors. In many neurological diseases, synapse structure and AMPA receptor function are altered, thus making AMPA receptors potential therapeutic targets for clinical intervention. The work summarized in this review suggests a link between AMPA receptor function and debilitating neuropathologies, and discusses the current state of therapies targeting AMPA receptors in four diseases. In amyotrophic lateral sclerosis, AMPA receptors allow cytotoxic levels of calcium into neurons, leading to motor neuron death. Likewise, in some epilepsies, overactivation of AMPA receptors leads to neuron damage. The same is true for ischemia, where oxygen deprivation leads to excitotoxicity. Conversely, Alzheimer's disease is characterized by decreased AMPA activation and synapse loss. Unfortunately, many clinical studies have had limited success by directly targeting AMPA receptors in these diseases. We also discuss how the use of AMPA receptor modulators, commonly known as ampakines, in neurological diseases initially seemed promising in animal studies, but mostly ineffective in clinical trials. We propose that indirectly affecting AMPA receptors, such as by modulating transmembrane AMPA receptor regulatory proteins or, more generally, by regulating glutamatergic transmission, may provide new therapeutic potential for neurological disorders.


Subject(s)
Nervous System Diseases/drug therapy , Receptors, AMPA/antagonists & inhibitors , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Epilepsy/drug therapy , Epilepsy/physiopathology , Excitatory Postsynaptic Potentials/drug effects , Humans , Mice , Molecular Targeted Therapy/trends , Neurotransmitter Agents/pharmacology , Neurotransmitter Agents/therapeutic use , Rats , Receptors, AMPA/agonists , Receptors, AMPA/physiology
13.
Glia ; 60(7): 1067-77, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22488940

ABSTRACT

Accumulating evidence supports the idea that synapses are tripartite, whereby perisynaptic astrocytes modulate both pre- and postsynaptic function. Although some of these features have been uncovered by using electrophysiological methods, less is known about the structural interplay between synapses and glial processes. Here, we investigated how astrocytes govern the plasticity of individual hippocampal dendritic spines. Recently, we uncovered that a subgroup of innervated dendritic spines is able to undergo remodeling by extending spine head protrusions (SHPs) toward neighboring functional presynaptic boutons, resulting in new synapses. Although glutamate serves as a trigger, how this behavior is regulated is unknown. As astrocytes control extracellular glutamate levels through their high-affinity uptake transporters, together with their privileged access to synapses, we investigated a role for astrocytes in SHP formation. Using time-lapse confocal microscopy, we found that the volume overlap between spines and astrocytic processes decreased during the formation of SHPs. Focal application of glutamate also reduced spine-astrocyte overlap and induced SHPs. Importantly, SHP formation was prevented by blocking glial glutamate transporters, suggesting that glial control of extracellular glutamate is important for SHP-mediated plasticity of spines. Hence, the dynamic changes of both spines and astrocytes can rapidly modify synaptic connectivity.


Subject(s)
Amino Acid Transport System X-AG/metabolism , Dendritic Spines/metabolism , Hippocampus/metabolism , Neuroglia/metabolism , Synapses/metabolism , Animals , Female , Glutamic Acid/metabolism , Male , Mice , Microscopy, Confocal , Neuronal Plasticity/physiology , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...