Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis ; 44(1): 43-8, 2015.
Article in English | MEDLINE | ID: mdl-25190626

ABSTRACT

Metabolic dysfunction exacerbates Alzheimer's disease (AD) incidence and progression. Here we report that activation of the AMPK pathway, a common target in the management of diabetes, results in gender-divergent cognitive effects in a murine model of the disease. Specifically, our results show that activation of AMPK increases memory dysfunction in males but is protective in females, suggesting that gender considerations may constitute an important factor in medical intervention of diabetes as well as AD.


Subject(s)
Alzheimer Disease/complications , Hypoglycemic Agents/therapeutic use , Memory Disorders/drug therapy , Memory Disorders/etiology , Metformin/therapeutic use , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Analysis of Variance , Animals , Blood Glucose/drug effects , Disease Models, Animal , Fasting , Humans , Maze Learning/drug effects , Memory Disorders/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics
2.
Proc Natl Acad Sci U S A ; 111(32): E3343-52, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25071179

ABSTRACT

Glial cells are an integral part of functional communication in the brain. Here we show that astrocytes contribute to the fast dynamics of neural circuits that underlie normal cognitive behaviors. In particular, we found that the selective expression of tetanus neurotoxin (TeNT) in astrocytes significantly reduced the duration of carbachol-induced gamma oscillations in hippocampal slices. These data prompted us to develop a novel transgenic mouse model, specifically with inducible tetanus toxin expression in astrocytes. In this in vivo model, we found evidence of a marked decrease in electroencephalographic (EEG) power in the gamma frequency range in awake-behaving mice, whereas neuronal synaptic activity remained intact. The reduction in cortical gamma oscillations was accompanied by impaired behavioral performance in the novel object recognition test, whereas other forms of memory, including working memory and fear conditioning, remained unchanged. These results support a key role for gamma oscillations in recognition memory. Both EEG alterations and behavioral deficits in novel object recognition were reversed by suppression of tetanus toxin expression. These data reveal an unexpected role for astrocytes as essential contributors to information processing and cognitive behavior.


Subject(s)
Astrocytes/physiology , Recognition, Psychology/physiology , Animals , Astrocytes/drug effects , Brain Waves/drug effects , Brain Waves/physiology , Calcium Signaling , Carbachol/pharmacology , Electroencephalography , Gene Expression , Glutamic Acid/metabolism , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Neurological , Nerve Net/cytology , Nerve Net/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Synaptic Transmission , Tetanus Toxin/genetics , Tetanus Toxin/metabolism , Tissue Culture Techniques , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
3.
Proc Natl Acad Sci U S A ; 110(27): E2518-27, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23776240

ABSTRACT

Synaptic loss is the cardinal feature linking neuropathology to cognitive decline in Alzheimer's disease (AD). However, the mechanism of synaptic damage remains incompletely understood. Here, using FRET-based glutamate sensor imaging, we show that amyloid-ß peptide (Aß) engages α7 nicotinic acetylcholine receptors to induce release of astrocytic glutamate, which in turn activates extrasynaptic NMDA receptors (eNMDARs) on neurons. In hippocampal autapses, this eNMDAR activity is followed by reduction in evoked and miniature excitatory postsynaptic currents (mEPSCs). Decreased mEPSC frequency may reflect early synaptic injury because of concurrent eNMDAR-mediated NO production, tau phosphorylation, and caspase-3 activation, each of which is implicated in spine loss. In hippocampal slices, oligomeric Aß induces eNMDAR-mediated synaptic depression. In AD-transgenic mice compared with wild type, whole-cell recordings revealed excessive tonic eNMDAR activity accompanied by eNMDAR-sensitive loss of mEPSCs. Importantly, the improved NMDAR antagonist NitroMemantine, which selectively inhibits extrasynaptic over physiological synaptic NMDAR activity, protects synapses from Aß-induced damage both in vitro and in vivo.


Subject(s)
Amyloid beta-Peptides/toxicity , Astrocytes/metabolism , Glutamic Acid/metabolism , Neural Inhibition/physiology , Peptide Fragments/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Astrocytes/pathology , Coculture Techniques , Female , Fluorescence Resonance Energy Transfer , HEK293 Cells , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Transgenic , Rats , Receptors, Nicotinic/metabolism , Synapses/metabolism , alpha7 Nicotinic Acetylcholine Receptor
4.
J Neurosci ; 33(4): 1314-25, 2013 Jan 23.
Article in English | MEDLINE | ID: mdl-23345207

ABSTRACT

Synapses onto distal dendritic tufts are believed to function by modulating time-locked proximal inputs; however, the role of these synapses when proximal inputs are asynchronous or silent is unknown. Surprisingly, we found that activation of apical tuft synapses alone resulted in heterosynaptic potentiation of proximal synapses. In mouse adult hippocampal CA1 pyramidal neurons, we show that activation of distal inputs from the entorhinal cortex (EC) specifically strengthens proximal synapses projecting from CA3. This slow AMPA receptor-mediated potentiation is accompanied by increased synaptic GluN2B-containing NMDA receptors, which are normally restricted to juvenile animals. These two synaptic modifications interact to generate striking bidirectional metaplastic changes. Heterosynaptically potentiated synapses become resistant to subsequent long-term potentiation (LTP) as the two forms of AMPA receptor-mediated potentiation occlude. However, this is only true when the LTP induction protocol is relatively weak. When it is strong and repeated, the magnitude of LTP after heterosynaptic plasticity is greatly increased, specifically through the activation of GluN2B-containing NMDA receptors. Thus, CA1 neurons expressing heterosynaptic potentiation induced by external sensory input from the EC become more strongly driven by internally generated environmental representations from CA3. Furthermore, subsequent SC LTP in this ensemble is shifted to potentiate only strongly activated CA3 inputs, while endowing these synapses with enhanced potentiation. These results show that one set of inputs can exert long-lasting heterosynaptic control over another, allowing the coupling of two functionally and spatially distinct pathways, thereby greatly expanding the repertoire of cellular and network plasticity.


Subject(s)
Dendrites/physiology , Long-Term Potentiation/physiology , Neural Pathways/physiology , Pyramidal Cells/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , CA1 Region, Hippocampal/physiology , Excitatory Postsynaptic Potentials/physiology , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Patch-Clamp Techniques
5.
J Neurosci ; 31(30): 10891-902, 2011 Jul 27.
Article in English | MEDLINE | ID: mdl-21795541

ABSTRACT

Nicotine is the primary psychoactive substance in tobacco, and it exerts its effects by interaction with various subtypes of nicotinic acetylcholine receptors (nAChRs) in the brain. One of the major subtypes expressed in brain, the α4ß2-nAChR, endogenously modulates neuronal excitability and thereby, modifies certain normal as well as nicotine-induced behaviors. Although α4-containing nAChRs are widely expressed across the brain, a major focus has been on their roles within midbrain dopaminergic regions involved in drug addiction, mental illness, and movement control in humans. We developed a unique model system to examine the role of α4-nAChRs within dopaminergic neurons by a targeted genetic deletion of the α4 subunit from dopaminergic neurons in mice. The loss α4 mRNA and α4ß2-nAChRs from dopaminergic neurons was confirmed, as well as selective loss of α4ß2-nAChR function from dopaminergic but not GABAergic neurons. Two behaviors central to nicotine dependence, reward and anxiety relief, were examined. α4-nAChRs specifically on dopaminergic neurons were demonstrated to be necessary for nicotine reward as measured by nicotine place preference, but not for another drug of addiction, cocaine. α4-nAChRs are necessary for the anxiolytic effects of nicotine in the elevated plus maze, and elimination of α4ß2-nAChRs specifically from dopaminergic neurons decreased sensitivity to the anxiolytic effects of nicotine. Deletion of α4-nAChRs specifically from dopaminergic neurons also increased sensitivity to nicotine-induced locomotor depression; however, nicotine-induced hypothermia was unaffected. This is the first work to develop a dopaminergic specific deletion of a nAChR subunit and examine resulting changes in nicotine-related behaviors.


Subject(s)
Anxiety/drug therapy , Dopamine/metabolism , Neurons/physiology , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Receptors, Nicotinic/metabolism , Reward , Analysis of Variance , Animals , Anxiety/pathology , Behavior, Animal , Body Temperature/drug effects , Body Temperature/genetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Disease Models, Animal , Dopamine Plasma Membrane Transport Proteins/genetics , Female , Glutamate Decarboxylase/metabolism , Male , Mesencephalon/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotinic Agonists/pharmacokinetics , Protein Binding/drug effects , Pyridines/pharmacokinetics , Receptors, Nicotinic/deficiency , Synaptosomes/drug effects , Synaptosomes/metabolism , Tritium/metabolism , gamma-Aminobutyric Acid/metabolism
6.
PLoS One ; 5(9)2010 Sep 29.
Article in English | MEDLINE | ID: mdl-20927382

ABSTRACT

A central concept in the field of learning and memory is that NMDARs are essential for synaptic plasticity and memory formation. Surprisingly then, multiple studies have found that behavioral experience can reduce or eliminate the contribution of these receptors to learning. The cellular mechanisms that mediate learning in the absence of NMDAR activation are currently unknown. To address this issue, we examined the contribution of Ca(2+)-permeable AMPARs to learning and plasticity in the hippocampus. Mutant mice were engineered with a conditional genetic deletion of GluR2 in the CA1 region of the hippocampus (GluR2-cKO mice). Electrophysiology experiments in these animals revealed a novel form of long-term potentiation (LTP) that was independent of NMDARs and mediated by GluR2-lacking Ca(2+)-permeable AMPARs. Behavioral analyses found that GluR2-cKO mice were impaired on multiple hippocampus-dependent learning tasks that required NMDAR activation. This suggests that AMPAR-mediated LTP interferes with NMDAR-dependent plasticity. In contrast, NMDAR-independent learning was normal in knockout mice and required the activation of Ca(2+)-permeable AMPARs. These results suggest that GluR2-lacking AMPARs play a functional and previously unidentified role in learning; they appear to mediate changes in synaptic strength that occur after plasticity has been established by NMDARs.


Subject(s)
Calcium/metabolism , Learning , Mice/physiology , Neuronal Plasticity , Receptors, AMPA/metabolism , Synapses/physiology , Animals , Female , Hippocampus/physiology , Long-Term Potentiation , Male , Mice/genetics , Mice, Knockout , Receptors, AMPA/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
7.
J Neurosci ; 30(34): 11501-5, 2010 Aug 25.
Article in English | MEDLINE | ID: mdl-20739572

ABSTRACT

NMDA receptors are typically excited by a combination of glutamate and glycine. Here we describe excitatory responses in CNS myelin that are gated by a glycine agonist alone and mediated by NR1/NR3 "NMDA" receptor subunits. Response properties include activation by d-serine, inhibition by the glycine-site antagonist CNQX, and insensitivity to the glutamate-site antagonist d-APV. d-Serine responses were abrogated in NR3A-deficient mice. Our results suggest the presence of functional NR1/NR3 receptors in CNS myelin.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Glycine/physiology , Myelin Sheath/physiology , Protein Subunits/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Cell Line , Central Nervous System/physiology , Humans , Mice , Mice, Knockout , Protein Subunits/agonists , Protein Subunits/genetics , Rats , Rats, Long-Evans , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/agonists , Recombinant Proteins/pharmacology
8.
J Neurosci ; 30(24): 8203-13, 2010 Jun 16.
Article in English | MEDLINE | ID: mdl-20554871

ABSTRACT

The central nucleus of the amygdala (CeA) has been identified as a site of nociceptive processing important for sensitization induced by peripheral injury. However, the cellular signaling components underlying this function remain unknown. Here, we identify metabotropic glutamate receptor 5 (mGluR5) as an integral component of nociceptive processing in the CeA. Pharmacological activation of mGluRs with (R,S)-3,5-dihydroxyphenylglycine (DHPG) in the CeA of mice is sufficient to induce peripheral hypersensitivity in the absence of injury. DHPG-induced peripheral hypersensitivity is reduced via pharmacological blockade of mGluR5 or genetic disruption of mGluR5. Furthermore, pharmacological blockade or conditional deletion of mGluR5 in the CeA abrogates inflammation-induced hypersensitivity, demonstrating the necessity of mGluR5 in CeA-mediated pain modulation. Moreover, we demonstrate that phosphorylation of extracellular-signal regulated kinase 1/2 (ERK1/2) is downstream of mGluR5 activation in the CeA and is necessary for the full expression of peripheral inflammation-induced behavioral sensitization. Finally, we present evidence of right hemispheric lateralization of mGluR5 modulation of amygdalar nociceptive processing. We demonstrate that unilateral pharmacological activation of mGluR5 in the CeA produces distinct behavioral responses depending on whether the right or left amygdala is injected. We also demonstrate significantly higher levels of mGluR5 expression in the right amygdala compared with the left under baseline conditions, suggesting a potential mechanism for right hemispheric lateralization of amygdala function in pain processing. Together, these results establish an integral role for mGluR5 and ERK1/2 in nociceptive processing in the CeA.


Subject(s)
Amygdala/metabolism , Pain/physiopathology , Receptors, Kainic Acid/metabolism , Amygdala/drug effects , Analysis of Variance , Animals , Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Formaldehyde , Functional Laterality , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Hyperalgesia/metabolism , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Pain/chemically induced , Pain/genetics , Pain Measurement/methods , Pyridines/pharmacology , Receptors, Glucocorticoid/deficiency , Receptors, Kainic Acid/deficiency
9.
Commun Integr Biol ; 2(5): 418-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19907705

ABSTRACT

At nerve terminals, neurotransmitter release is mediated by exocytosis of synaptic vesicles at active zone. After exocytosis, vesicular components are efficiently retrieved by endocytosis. Tight coupling between synaptic vesicle exocytosis and endocytosis is critical for the maintenance of neurotransmission at central synapses. Recently, we have developed a new fluorescent pH reporter that permits us to examine exocytosis-endocytosis coupling at the level of individual synaptic vesicles at hippocampal synapses.1 To our surprise, we observed that the tight coupling of exocytosis and endocytosis broke down at very low stimulation frequencies, resulting in the generation of two endocytic vesicles per single exocytic fusion event. As stimulation frequency increased, exocytosis-endocytic coupling was restored with one endocytic vesicle generated for each vesicle exocytosed. Further studies revealed that the dissimilar patterns of exocytosis-endocytic coupling at different stimulation frequencies were mediated by two pathways of endocytosis that are orchestrated during differential patterns of neuronal activity.1 Here, we summarize our observations and further discuss their possible implications.

10.
J Neurosci ; 29(27): 8805-15, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19587288

ABSTRACT

It has been recently shown that the Alzheimer's disease (AD) pathogenic peptide amyloid beta(1-42) (Abeta(1-42)) binds to the alpha7 nicotinic acetylcholine receptor (alpha7nAChR) with high affinity and the alpha7nAChR and Abeta(1-42) are both found colocalized in neuritic plaques of human brains with AD. Moreover, the intraneuronal accumulation of Abeta(1-42) was shown to be facilitated by its high-affinity binding to the alpha7nAChR, and alpha7nAChR activation mediates Abeta-induced tau protein phosphorylation. To test the hypothesis that alpha7nAChRs are involved in AD pathogenesis, we used a transgenic mouse model of AD overexpressing a mutated form of the human amyloid precursor protein (APP) and lacking the alpha7nAChR gene (APPalpha7KO). We have shown that, despite the presence of high amounts of APP and amyloid deposits, deleting the alpha7nAChR subunit in the mouse model of AD leads to a protection from the dysfunction in synaptic integrity (pathology and plasticity) and learning and memory behavior. Specifically, APPalpha7KO mice express APP and Abeta at levels similar to APP mice, and yet they were able to solve a cognitive challenge such as the Morris water maze test significantly better than APP, with performances comparable to control groups. Moreover, deleting the alpha7nAChR subunit protected the brain from loss of the synaptic markers synaptophysin and MAP2, reduced the gliosis, and preserved the capacity to elicit long-term potentiation otherwise deficient in APP mice. These results are consistent with the hypothesis that the alpha7nAChR plays a role in AD and suggest that interrupting alpha7nAChR function could be beneficial in the treatment of AD.


Subject(s)
Alzheimer Disease/genetics , Cognition Disorders/genetics , Disease Models, Animal , Gene Deletion , Receptors, Nicotinic/genetics , Synapses/genetics , Animals , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, Nicotinic/deficiency , Serum Amyloid A Protein/biosynthesis , Serum Amyloid A Protein/genetics , alpha7 Nicotinic Acetylcholine Receptor
11.
Proc Natl Acad Sci U S A ; 106(16): 6766-71, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19342486

ABSTRACT

Mutations in doublecortin (DCX) are associated with intractable epilepsy in humans, due to a severe disorganization of the neocortex and hippocampus known as classical lissencephaly. However, the basis of the epilepsy in lissencephaly remains unclear. To address potential functional redundancy with murin Dcx, we targeted one of the closest homologues, doublecortin-like kinase 2 (Dclk2). Here, we report that Dcx; Dclk2-null mice display frequent spontaneous seizures that originate in the hippocampus, with most animals dying in the first few months of life. Elevated hippocampal expression of c-fos and loss of somatostatin-positive interneurons were identified, both known to correlate with epilepsy. Dcx and Dclk2 are coexpressed in developing hippocampus, and, in their absence, there is dosage-dependent disrupted hippocampal lamination associated with a cell-autonomous simplification of pyramidal dendritic arborizations leading to reduced inhibitory synaptic tone. These data suggest that hippocampal dysmaturation and insufficient receptive field for inhibitory input may underlie the epilepsy in lissencephaly, and suggest potential therapeutic strategies for controlling epilepsy in these patients.


Subject(s)
Cell Differentiation , Hippocampus/enzymology , Hippocampus/pathology , Microtubule-Associated Proteins/deficiency , Neurons/enzymology , Neuropeptides/deficiency , Protein Serine-Threonine Kinases/deficiency , Seizures/enzymology , Animals , Cell Differentiation/drug effects , Cell Polarity/drug effects , Dendrites/drug effects , Dendrites/pathology , Doublecortin Domain Proteins , Doublecortin Protein , Hippocampus/embryology , Interneurons/drug effects , Interneurons/enzymology , Interneurons/pathology , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Neuropeptides/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/enzymology , Pyramidal Cells/pathology , Seizures/pathology , Somatostatin/metabolism , Survival Analysis , Synapses/drug effects , Synapses/metabolism , Weaning , gamma-Aminobutyric Acid/pharmacology
12.
J Neurosci ; 29(12): 3676-84, 2009 Mar 25.
Article in English | MEDLINE | ID: mdl-19321764

ABSTRACT

The mechanisms that contribute to the extinction of previously acquired memories are not well understood. These processes, often referred to as inhibitory learning, are thought to be parallel learning mechanisms that require a reacquisition of new information and suppression of previously acquired experiences in order to adapt to novel situations. Using newly generated metabotropic glutamate receptor 5 (mGluR5) knock-out mice, we investigated the role of mGluR5 in the acquisition and reversal of an associative conditioned task and a spatial reference task. We found that acquisition of fear conditioning is partially impaired in mice lacking mGluR5. More markedly, we found that extinction of both contextual and auditory fear was completely abolished in mGluR5 knock-out mice. In the Morris Water Maze test (MWM), mGluR5 knock-out mice exhibited mild deficits in the rate of acquisition of the regular water maze task, but again had significant deficits in the reversal task, despite overall spatial memory being intact. Together, these results demonstrate that mGluR5 is critical to the function of neural circuits that are required for inhibitory learning mechanisms, and suggest that targeting metabotropic receptors may be useful in treating psychiatric disorders in which aversive memories are inappropriately retained.


Subject(s)
Association Learning , Avoidance Learning , Extinction, Psychological , Maze Learning , Receptors, Metabotropic Glutamate/physiology , Acoustic Stimulation , Animals , Conditioning, Operant , Fear , Memory , Mice , Mice, Knockout , Receptor, Metabotropic Glutamate 5 , Receptors, Metabotropic Glutamate/genetics
13.
Neuron ; 61(3): 397-411, 2009 Feb 12.
Article in English | MEDLINE | ID: mdl-19217377

ABSTRACT

Synaptic vesicle recycling is essential for maintaining efficient synaptic transmission. Detailed dissection of single-vesicle recycling still remains a major challenge. We have developed a fluorescent pH reporter that permits us to follow the fate of individual vesicles at hippocampal synapses after exocytosis. Here we show that, during low-frequency stimulation, single-vesicle fusion leads to two distinct vesicle internalizations, instead of one, as in general perception: one by a fast endocytosis pathway ( approximately 3 s), the other by a slow endocytosis pathway (after 10 s). The exocytosed vesicular proteins are preferentially recaptured in both pathways. RNAi knockdown of clathrin inhibits both pathways. As stimulation frequency increases, the number of endocytosed vesicles begins to match antecedent exocytosis. Meanwhile, the slow endocytosis is accelerated and becomes the predominant pathway. These results reveal that two pathways of endocytosis are orchestrated during neuronal activity, establishing a highly efficient endocytosis at central synapses.


Subject(s)
Hippocampus/metabolism , Neurons/metabolism , Presynaptic Terminals/metabolism , Synaptic Vesicles/metabolism , Animals , Cells, Cultured , Clathrin/genetics , Endocytosis/physiology , Exocytosis/physiology , Fluorescent Dyes , Hippocampus/ultrastructure , Membrane Fusion/physiology , Microscopy, Fluorescence/methods , Neurons/ultrastructure , Presynaptic Terminals/ultrastructure , Pyridinium Compounds , Quaternary Ammonium Compounds , RNA Interference , Rats , Staining and Labeling/methods , Synaptic Transmission/physiology , Synaptic Vesicles/ultrastructure , Time Factors
14.
Proc Natl Acad Sci U S A ; 104(29): 12181-6, 2007 Jul 17.
Article in English | MEDLINE | ID: mdl-17620617

ABSTRACT

Presynaptic ionotropic glutamate receptors are emerging as key players in the regulation of synaptic transmission. Here we identify GluR7, a kainate receptor (KAR) subunit with no known function in the brain, as an essential subunit of presynaptic autoreceptors that facilitate hippocampal mossy fiber synaptic transmission. GluR7(-/-) mice display markedly reduced short- and long-term synaptic potentiation. Our data suggest that presynaptic KARs are GluR6/GluR7 heteromers that coassemble and are localized within synapses. We show that recombinant GluR6/GluR7 KARs exhibit low sensitivity to glutamate, and we provide evidence that presynaptic KARs at mossy fiber synapses are likely activated by high concentrations of glutamate. Overall, from our data, we propose a model whereby presynaptic KARs are localized in the presynaptic active zone close to release sites, display low affinity for glutamate, are likely Ca(2+)-permeable, are activated by single release events, and operate within a short time window to facilitate the subsequent release of glutamate.


Subject(s)
Autoreceptors/metabolism , Mossy Fibers, Hippocampal/metabolism , Presynaptic Terminals/metabolism , Receptors, Kainic Acid/metabolism , Animals , Cell Line , Excitatory Postsynaptic Potentials , Humans , Long-Term Potentiation , Mice , Protein Subunits/metabolism , Protein Transport , Receptors, Kainic Acid/deficiency , Recombinant Proteins/metabolism , Subcellular Fractions/metabolism , Time Factors , GluK2 Kainate Receptor , GluK3 Kainate Receptor
15.
Pharmacol Biochem Behav ; 87(1): 146-57, 2007 May.
Article in English | MEDLINE | ID: mdl-17509676

ABSTRACT

Nicotine, via a family of nicotinic acetylcholine receptors, elicits many physiological responses, including alterations in anxiety. Studies suggest that the effects of nicotine on anxiety may support smoking behaviors. We reported previously that mice lacking the beta3 nicotinic receptor subunit demonstrate increased activity in the open field arena. Open field activity has been shown to be a composite of anxiety and locomotor activity, behaviors that are both altered by nicotine. We therefore sought to differentiate the role(s) of beta3-containing receptors in anxiety and locomotor activity. Anxiety behaviors were examined in the elevated plus maze, the black/white box and the mirrored chamber. Beta3 null mutant mice demonstrated decreased anxiety with more time spent on the open arm of the elevated plus maze than their wildtype littermates. No significant differences were observed with the black/white box or the mirrored chamber. Levels of the stress hormone, corticosterone, were significantly higher in the beta3 null mutant mice at baseline and following exposure to stress. Increased locomotor activity in the Y-maze was also observed for the beta3 null mutant mice, but only following exposure to stress. These findings strongly suggest that beta3-containing nicotinic receptors influence anxiety and may be critical for the continuation of smoking behaviors.


Subject(s)
Anxiety/psychology , Receptors, Nicotinic/genetics , Animals , Corticosterone/blood , Memory/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Protein Subunits/genetics , Stress, Psychological/genetics , Stress, Psychological/psychology
16.
J Neurosci ; 26(52): 13428-36, 2006 Dec 27.
Article in English | MEDLINE | ID: mdl-17192425

ABSTRACT

Synaptic dysfunction has been shown to be one of the earliest correlates of disease progression in animal models of Alzheimer's disease. Amyloid-beta protein (Abeta) is thought to play an important role in disease-related synaptic dysfunction, but the mechanism by which Abeta leads to synaptic dysfunction is not understood. Here we describe evidence that cleavage of APP in the C terminus may be necessary for the deficits present in APP transgenic mice. In APP transgenic mice with a mutated cleavage site at amino acid 664, normal synaptic transmission, synaptic plasticity, and learning were maintained despite the presence of elevated levels of APP, Abeta42, and even plaque accumulation. These results indicate that cleavage of APP may play a critical role in the development of synaptic and behavioral dysfunction in APP transgenic mice.


Subject(s)
Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Behavior, Animal , Learning/physiology , Synaptic Transmission/genetics , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Animals , Behavior, Animal/physiology , Excitatory Postsynaptic Potentials/genetics , Female , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Mutagenesis, Site-Directed , Neuronal Plasticity/genetics , Peptide Fragments/deficiency , Peptide Fragments/genetics , Peptide Fragments/metabolism
17.
J Neurosci ; 26(38): 9780-93, 2006 Sep 20.
Article in English | MEDLINE | ID: mdl-16988049

ABSTRACT

Neuronal nicotinic acetylcholine receptors (nAChRs) belong to a family of ligand-gated ion channels that play important roles in central and peripheral nervous systems. The subcellular distribution of neuronal nAChRs has important implications for function and is not well understood. Here, we analyzed the targeting of two major types of neuronal nAChRs by expressing epitope-tagged subunits in cultured hippocampal neurons. Surprisingly, the alpha7 nAChR (alpha7) and alpha4/beta2 nAChR (alpha4beta2) displayed distinct patterns of expression, with alpha7 targeted preferentially to the somatodendritic compartments, whereas alpha4beta2 was localized to both axonal and dendritic domains. When fused to CD4 or IL2RA (interleukin 2 receptor alpha subunit) proteins, which are normally distributed ubiquitously, the M3-M4 intracellular loop from the alpha7 subunit promoted dendritic expression, whereas the homologous M3-M4 loop from the alpha4 subunit led to surface axonal expression. Systemic screening and alanine substitution further identified a 25-residue leucine motif ([DE]XXXL[LI]) containing an axonal targeting sequence within the alpha4 loop and a 48-residue dileucine and tyrosine motif (YXXØ) containing a dendritic targeting sequence from the alpha7 loop. These results provide valuable information in understanding diverse roles of neuronal nAChRs in mediating and modulating synaptic transmission, synaptic plasticity, and nicotine addiction.


Subject(s)
Axons/metabolism , Dendrites/metabolism , Gene Targeting/methods , Receptors, Nicotinic/chemistry , Receptors, Nicotinic/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Cells, Cultured , Hippocampus/metabolism , Humans , Molecular Sequence Data , Rats , Receptors, Nicotinic/biosynthesis
18.
Nat Neurosci ; 9(5): 611-21, 2006 May.
Article in English | MEDLINE | ID: mdl-16617342

ABSTRACT

A key step in glutamatergic synapse maturation is the replacement of developmentally expressed N-methyl-D-aspartate receptors (NMDARs) with mature forms that differ in subunit composition, electrophysiological properties and propensity to elicit synaptic plasticity. However, the mechanisms underlying the removal and replacement of synaptic NMDARs are poorly understood. Here we demonstrate that NMDARs containing the developmentally regulated NR3A subunit undergo rapid endocytosis from the dendritic plasma membrane in cultured rat hippocampal neurons. This endocytic removal is regulated by PACSIN1/syndapin1, which directly and selectively binds the carboxy-terminal domain of NR3A through its NPF motifs and assembles a complex of proteins including dynamin and clathrin. Endocytosis of NR3A by PACSIN1 is activity dependent, and disruption of PACSIN1 function causes NR3A accumulation at synaptic sites. Our results reveal a new activity-dependent mechanism involved in the regulation of NMDAR expression at synapses during development, and identify a brain-specific endocytic adaptor that confers spatiotemporal and subunit specificity to NMDAR endocytosis.


Subject(s)
Endocytosis/physiology , Membrane Glycoproteins/physiology , Neurons/cytology , Proteoglycans/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Animals , Blotting, Western/methods , Cells, Cultured , Cloning, Molecular/methods , Electrophysiology/methods , Embryo, Mammalian , Endocytosis/drug effects , Fluorescent Antibody Technique/methods , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Humans , In Situ Hybridization/methods , Membrane Glycoproteins/genetics , Microscopy, Immunoelectron/methods , Mutation/physiology , N-Methylaspartate/pharmacology , Neurons/drug effects , Proteoglycans/genetics , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Synapses/drug effects , Synapses/ultrastructure , Syndecans , Transfection/methods , Two-Hybrid System Techniques
19.
J Neurosci ; 26(13): 3474-81, 2006 Mar 29.
Article in English | MEDLINE | ID: mdl-16571754

ABSTRACT

The mossy fiber to CA3 pyramidal neuron synapse in the hippocampus displays an atypical form of NMDA receptor-independent long-term potentiation (LTP). Plasticity at this synapse is expressed in the presynaptic terminal as an elevated probability of neurotransmitter release. However, evidence indicates that postsynaptic mechanisms and trans-synaptic signaling through an association between postsynaptic EphB receptors and presynaptic B-ephrins are necessary for the induction of LTP. Here we show that ephrin-B3 protein is highly expressed in mossy fiber axons and terminals. There are specific deficits in mossy fiber LTP in mice in which the cytoplasmic C-terminal signaling domain of the ephrin-B3 protein is replaced with beta-galactosidase. These deficits are not observed in ephrin-B3 null mutant mice because of functional redundancy by virtue of other B-ephrins. These results indicate that B-ephrin reverse signaling into the presynaptic mossy fiber bouton is required for the induction of NMDA receptor-independent LTP at this synapse.


Subject(s)
Ephrin-B3/metabolism , Long-Term Potentiation/physiology , Mossy Fibers, Hippocampal/physiology , Pyramidal Cells/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/physiology , Synaptic Transmission/physiology , Animals , Cells, Cultured , Mice , N-Methylaspartate/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...