Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
Biol Psychiatry ; 91(8): 740-752, 2022 04 15.
Article in English | MEDLINE | ID: mdl-34952697

ABSTRACT

BACKGROUND: NMDA receptor (NMDAR) hypofunction has been implicated in several psychiatric disorders with impairment of cognitive flexibility. However, the molecular mechanism of how NMDAR hypofunction with decreased NMDAR tone causes the impairment of cognitive flexibility has been minimally understood. Furthermore, it has been unclear whether hippocampal astrocytes regulate NMDAR tone and cognitive flexibility. METHODS: We employed cell type-specific genetic manipulations, ex vivo electrophysiological recordings, sniffer patch recordings, cutting-edge biosensor for norepinephrine, and behavioral assays to investigate whether astrocytes can regulate NMDAR tone by releasing D-serine and glutamate. Subsequently, we further investigated the role of NMDAR tone in heterosynaptic long-term depression, metaplasticity, and cognitive flexibility. RESULTS: We found that hippocampal astrocytes regulate NMDAR tone via BEST1-mediated corelease of D-serine and glutamate. Best1 knockout mice exhibited reduced NMDAR tone and impairments of homosynaptic and α1 adrenergic receptor-dependent heterosynaptic long-term depression, which leads to defects in metaplasticity and cognitive flexibility. These impairments in Best1 knockout mice can be rescued by hippocampal astrocyte-specific BEST1 expression or enhanced NMDAR tone through D-serine supplement. D-serine injection in Best1 knockout mice during initial learning rescues subsequent reversal learning. CONCLUSIONS: These findings indicate that NMDAR tone during initial learning is important for subsequent learning, and hippocampal NMDAR tone regulated by astrocytic BEST1 is critical for heterosynaptic long-term depression, metaplasticity, and cognitive flexibility.


Subject(s)
Astrocytes , Receptors, N-Methyl-D-Aspartate , Animals , Astrocytes/metabolism , Bestrophins/metabolism , Glutamic Acid/metabolism , Hippocampus/metabolism , Humans , Mice , Receptors, N-Methyl-D-Aspartate/physiology , Serine/metabolism
2.
Exp Neurobiol ; 24(1): 17-23, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25792866

ABSTRACT

Astrocytes and neurons are inseparable partners in the brain. Neurotransmitters released from neurons activate corresponding G protein-coupled receptors (GPCR) expressed in astrocytes, resulting in release of gliotransmitters such as glutamate, D-serine, and ATP. These gliotransmitters in turn influence neuronal excitability and synaptic activities. Among these gliotransmitters, ATP regulates the level of network excitability and is critically involved in sleep homeostasis and astrocytic Ca(2+) oscillations. ATP is known to be released from astrocytes by Ca(2+)-dependent manner. However, the precise source of Ca(2+), whether it is Ca(2+) entry from outside of cell or from the intracellular store, is still not clear yet. Here, we performed sniffer patch to detect ATP release from astrocyte by using various stimulation. We found that ATP was not released from astrocyte when Ca(2+) was released from intracellular stores by activation of Gαq-coupled GPCR including PAR1, P2YR, and B2R. More importantly, mechanical stimulation (MS)-induced ATP release from astrocyte was eliminated when external Ca(2+) was omitted. Our results suggest that Ca(2+) entry, but not release from intracellular Ca(2+) store, is critical for MS-induced ATP release from astrocyte.

3.
J Physiol ; 592(22): 4951-68, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25239459

ABSTRACT

GABA is the major inhibitory transmitter in the brain and is released not only from a subset of neurons but also from glia. Although neuronal GABA is well known to be synthesized by glutamic acid decarboxylase (GAD), the source of glial GABA is unknown. After estimating the concentration of GABA in Bergmann glia to be around 5-10 mM by immunogold electron microscopy, we demonstrate that GABA production in glia requires MAOB, a key enzyme in the putrescine degradation pathway. In cultured cerebellar glia, both Ca(2+)-induced and tonic GABA release are significantly reduced by both gene silencing of MAOB and the MAOB inhibitor selegiline. In the cerebellum and striatum of adult mice, general gene silencing, knock out of MAOB or selegiline treatment resulted in elimination of tonic GABA currents recorded from granule neurons and medium spiny neurons. Glial-specific rescue of MAOB resulted in complete rescue of tonic GABA currents. Our results identify MAOB as a key synthesizing enzyme of glial GABA, which is released via bestrophin 1 (Best1) channel to mediate tonic inhibition in the brain.


Subject(s)
Monoamine Oxidase/metabolism , Neuroglia/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Cerebellum/cytology , Cerebellum/metabolism , Corpus Striatum/cytology , Corpus Striatum/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Monoamine Oxidase/genetics , Neural Inhibition , Neuroglia/physiology
4.
Nat Med ; 20(8): 886-96, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24973918

ABSTRACT

In Alzheimer's disease (AD), memory impairment is the most prominent feature that afflicts patients and their families. Although reactive astrocytes have been observed around amyloid plaques since the disease was first described, their role in memory impairment has been poorly understood. Here, we show that reactive astrocytes aberrantly and abundantly produce the inhibitory gliotransmitter GABA by monoamine oxidase-B (Maob) and abnormally release GABA through the bestrophin 1 channel. In the dentate gyrus of mouse models of AD, the released GABA reduces spike probability of granule cells by acting on presynaptic GABA receptors. Suppressing GABA production or release from reactive astrocytes fully restores the impaired spike probability, synaptic plasticity, and learning and memory in the mice. In the postmortem brain of individuals with AD, astrocytic GABA and MAOB are significantly upregulated. We propose that selective inhibition of astrocytic GABA synthesis or release may serve as an effective therapeutic strategy for treating memory impairment in AD.


Subject(s)
Alzheimer Disease/metabolism , Astrocytes/metabolism , Eye Proteins/metabolism , Ion Channels/metabolism , Memory Disorders/metabolism , Monoamine Oxidase/metabolism , gamma-Aminobutyric Acid/biosynthesis , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Astrocytes/drug effects , Bestrophins , Cerebellum/metabolism , Dentate Gyrus/metabolism , Disease Models, Animal , Eye Proteins/genetics , GABA Antagonists/therapeutic use , Hippocampus/cytology , Hippocampus/metabolism , Humans , Ion Channels/genetics , Male , Maze Learning , Memory/drug effects , Memory Disorders/drug therapy , Memory Disorders/etiology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic , Monoamine Oxidase/genetics , Monoamine Oxidase Inhibitors/pharmacology , Peptide Fragments/pharmacology , Plaque, Amyloid/metabolism , Putrescine/pharmacology , RNA, Small Interfering/genetics , Receptors, GABA/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL