Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
EMBO Rep ; 23(10): e54420, 2022 10 06.
Article in English | MEDLINE | ID: mdl-35969184

ABSTRACT

Bipolar disorder (BD) is a chronic mood disorder characterized by manic and depressive episodes. Dysregulation of neuroplasticity and calcium homeostasis are frequently observed in BD patients, but the underlying molecular mechanisms are largely unknown. Here, we show that miR-499-5p regulates dendritogenesis and cognitive function by downregulating the BD risk gene CACNB2. miR-499-5p expression is increased in peripheral blood of BD patients, as well as in the hippocampus of rats which underwent juvenile social isolation. In rat hippocampal neurons, miR-499-5p impairs dendritogenesis and reduces surface expression and activity of the L-type calcium channel Cav1.2. We further identified CACNB2, which encodes a regulatory ß-subunit of Cav1.2, as a direct functional target of miR-499-5p in neurons. miR-499-5p overexpression in the hippocampus in vivo induces short-term memory impairments selectively in rats haploinsufficient for the Cav1.2 pore forming subunit Cacna1c. In humans, miR-499-5p expression is negatively associated with gray matter volumes of the left superior temporal gyrus, a region implicated in auditory and emotional processing. We propose that stress-induced miR-499-5p overexpression contributes to dendritic impairments, deregulated calcium homeostasis, and neurocognitive dysfunction in BD.


Subject(s)
Bipolar Disorder , Calcium Channels, L-Type , MicroRNAs , Animals , Bipolar Disorder/genetics , Bipolar Disorder/metabolism , Calcium/metabolism , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Hippocampus/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neuronal Plasticity/genetics , Rats
2.
Elife ; 112022 03 15.
Article in English | MEDLINE | ID: mdl-35290180

ABSTRACT

The proper development and function of neuronal circuits rely on a tightly regulated balance between excitatory and inhibitory (E/I) synaptic transmission, and disrupting this balance can cause neurodevelopmental disorders, for example, schizophrenia. MicroRNA-dependent gene regulation in pyramidal neurons is important for excitatory synaptic function and cognition, but its role in inhibitory interneurons is poorly understood. Here, we identify miR138-5p as a regulator of short-term memory and inhibitory synaptic transmission in the mouse hippocampus. Sponge-mediated miR138-5p inactivation specifically in mouse parvalbumin (PV)-expressing interneurons impairs spatial recognition memory and enhances GABAergic synaptic input onto pyramidal neurons. Cellular and behavioral phenotypes associated with miR138-5p inactivation are paralleled by an upregulation of the schizophrenia (SCZ)-associated Erbb4, which we validated as a direct miR138-5p target gene. Our findings suggest that miR138-5p is a critical regulator of PV interneuron function in mice, with implications for cognition and SCZ. More generally, they provide evidence that microRNAs orchestrate neural circuit development by fine-tuning both excitatory and inhibitory synaptic transmission.


Subject(s)
Memory, Short-Term , MicroRNAs , Animals , Hippocampus/physiology , Interneurons/physiology , Mice , MicroRNAs/genetics , Parvalbumins/metabolism
3.
EMBO Rep ; 22(10): e52094, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34396684

ABSTRACT

Synaptic scaling is a form of homeostatic plasticity which allows neurons to adjust their action potential firing rate in response to chronic alterations in neural activity. Synaptic scaling requires profound changes in gene expression, but the relative contribution of local and cell-wide mechanisms is controversial. Here we perform a comprehensive multi-omics characterization of the somatic and process compartments of primary rat hippocampal neurons during synaptic scaling. We uncover both highly compartment-specific and correlating changes in the neuronal transcriptome and proteome. Whereas downregulation of crucial regulators of neuronal excitability occurs primarily in the somatic compartment, structural components of excitatory postsynapses are mostly downregulated in processes. Local inhibition of protein synthesis in processes during scaling is confirmed for candidate synaptic proteins. Motif analysis further suggests an important role for trans-acting post-transcriptional regulators, including RNA-binding proteins and microRNAs, in the local regulation of the corresponding mRNAs. Altogether, our study indicates that, during synaptic scaling, compartmentalized gene expression changes might co-exist with neuron-wide mechanisms to allow synaptic computation and homeostasis.


Subject(s)
Neuronal Plasticity , Synapses , Animals , Gene Expression , Gene Expression Regulation , Neuronal Plasticity/genetics , Neurons , Rats
4.
EMBO Rep ; 20(2)2019 02.
Article in English | MEDLINE | ID: mdl-30552145

ABSTRACT

Aberrant synaptic function is thought to underlie social deficits in neurodevelopmental disorders such as autism and schizophrenia. Although microRNAs have been shown to regulate synapse development and plasticity, their potential involvement in the control of social behaviour in mammals remains unexplored. Here, we show that deletion of the placental mammal-specific miR379-410 cluster in mice leads to hypersocial behaviour, which is accompanied by increased excitatory synaptic transmission, and exaggerated expression of ionotropic glutamate receptor complexes in the hippocampus. Bioinformatic analyses further allowed us to identify five "hub" microRNAs whose deletion accounts largely for the upregulation of excitatory synaptic genes observed, including Cnih2, Dlgap3, Prr7 and Src. Thus, the miR379-410 cluster acts a natural brake for sociability, and interfering with specific members of this cluster could represent a therapeutic strategy for the treatment of social deficits in neurodevelopmental disorders.


Subject(s)
Behavior, Animal , Eutheria/genetics , MicroRNAs/genetics , Multigene Family , Social Behavior , Animals , Binding Sites , Eutheria/metabolism , Excitatory Postsynaptic Potentials , Genetic Association Studies , Genetic Markers , Hippocampus/metabolism , Mice , Mice, Knockout , Phenotype , Protein Interaction Mapping , Protein Interaction Maps , Pyramidal Cells/metabolism , RNA Interference , Receptors, Glutamate/metabolism , Synaptic Transmission
5.
Front Mol Neurosci ; 11: 171, 2018.
Article in English | MEDLINE | ID: mdl-29942249

ABSTRACT

microRNAs (miRNAs) have emerged as critical regulators of neuronal dendrite development. Specific precursor (pre-)miRNAs are actively transported to dendrites, but whether this process is regulated by neuronal activity and involved in activity-dependent dendritogenesis is unknown. Here we show that BDNF, a neurotrophin that is released in response to increased neuronal activity, promotes dendritic accumulation of pre-miR-134. Dendritic accumulation, but not transcription of pre-miR-134, is abrogated by treatment of neurons with the NMDA receptor (NMDAR) antagonist APV. Furthermore, APV interferes with BDNF-mediated repression of the known miR-134 target Pumilio 2 (Pum2) in a miR-134 binding site-specific manner. At the functional level, both APV treatment and knockdown of the pre-miR-134 transport protein DHX36 antagonize BDNF-induced dendritogenesis. These effects are likely mediated by reduced dendritic miR-134 activity, since both transfection of a synthetic miR-134 duplex or of a dendritically targeted pre-miR-134-181a chimera rescues BDNF-dependent dendritogenesis in the presence of APV. In conclusion, we have identified a novel NMDAR-dependent mechanism involved in the activity-dependent control of miRNA function during neuronal development.

6.
EMBO Rep ; 19(3)2018 03.
Article in English | MEDLINE | ID: mdl-29263199

ABSTRACT

MicroRNAs are important regulators of local protein synthesis during neuronal development. We investigated the dynamic regulation of microRNA production and found that the majority of the microRNA-generating complex, consisting of Dicer, TRBP, and PACT, specifically associates with intracellular membranes in developing neurons. Stimulation with brain-derived neurotrophic factor (BDNF), which promotes dendritogenesis, caused the redistribution of TRBP from the endoplasmic reticulum into the cytoplasm, and its dissociation from Dicer, in a Ca2+-dependent manner. As a result, the processing of a subset of neuronal precursor microRNAs, among them the dendritically localized pre-miR16, was impaired. Decreased production of miR-16-5p, which targeted the BDNF mRNA itself, was rescued by expression of a membrane-targeted TRBP Moreover, miR-16-5p or membrane-targeted TRBP expression blocked BDNF-induced dendritogenesis, demonstrating the importance of neuronal TRBP dynamics for activity-dependent neuronal development. We propose that neurons employ specialized mechanisms to modulate local gene expression in dendrites, via the dynamic regulation of microRNA biogenesis factors at intracellular membranes of the endoplasmic reticulum, which in turn is crucial for neuronal dendrite complexity and therefore neuronal circuit formation and function.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Dendrites/genetics , MicroRNAs/genetics , Neurogenesis/genetics , Nuclear Receptor Coactivators/genetics , Animals , DEAD-box RNA Helicases/genetics , Embryo, Mammalian , Embryonic Development/genetics , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Humans , Neurons/metabolism , RNA-Binding Proteins/genetics , Rats , Ribonuclease III/genetics
7.
EMBO J ; 36(12): 1770-1787, 2017 06 14.
Article in English | MEDLINE | ID: mdl-28487411

ABSTRACT

Synaptic downscaling is a homeostatic mechanism that allows neurons to reduce firing rates during chronically elevated network activity. Although synaptic downscaling is important in neural circuit development and epilepsy, the underlying mechanisms are poorly described. We performed small RNA profiling in picrotoxin (PTX)-treated hippocampal neurons, a model of synaptic downscaling. Thereby, we identified eight microRNAs (miRNAs) that were increased in response to PTX, including miR-129-5p, whose inhibition blocked synaptic downscaling in vitro and reduced epileptic seizure severity in vivo Using transcriptome, proteome, and bioinformatic analysis, we identified the calcium pump Atp2b4 and doublecortin (Dcx) as miR-129-5p targets. Restoring Atp2b4 and Dcx expression was sufficient to prevent synaptic downscaling in PTX-treated neurons. Furthermore, we characterized a functional crosstalk between miR-129-5p and the RNA-binding protein (RBP) Rbfox1. In the absence of PTX, Rbfox1 promoted the expression of Atp2b4 and Dcx. Upon PTX treatment, Rbfox1 expression was downregulated by miR-129-5p, thereby allowing the repression of Atp2b4 and Dcx. We therefore identified a novel activity-dependent miRNA/RBP crosstalk during synaptic scaling, with potential implications for neural network homeostasis and epileptogenesis.


Subject(s)
Gene Expression Regulation , MicroRNAs/metabolism , RNA Splicing Factors/metabolism , Synapses/physiology , Animals , Computational Biology , Doublecortin Domain Proteins , Doublecortin Protein , Gene Expression Profiling , Hippocampus/drug effects , Hippocampus/physiology , Mice , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Picrotoxin/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Proteome/analysis
8.
9.
Nat Neurosci ; 18(5): 666-73, 2015 May.
Article in English | MEDLINE | ID: mdl-25867122

ABSTRACT

The E3 ubiquitin ligase Ube3a is an important regulator of activity-dependent synapse development and plasticity. Ube3a mutations cause Angelman syndrome and have been associated with autism spectrum disorders (ASD). However, the biological significance of alternative Ube3a transcripts generated in mammalian neurons remains unknown. We report here that Ube3a1 RNA, a transcript that encodes a truncated Ube3a protein lacking catalytic activity, prevents exuberant dendrite growth and promotes spine maturation in rat hippocampal neurons. Surprisingly, Ube3a1 RNA function was independent of its coding sequence but instead required a unique 3' untranslated region and an intact microRNA pathway. Ube3a1 RNA knockdown increased activity of the plasticity-regulating miR-134, suggesting that Ube3a1 RNA acts as a dendritic competing endogenous RNA. Accordingly, the dendrite-growth-promoting effect of Ube3a1 RNA knockdown in vivo is abolished in mice lacking miR-134. Taken together, our results define a noncoding function of an alternative Ube3a transcript in dendritic protein synthesis, with potential implications for Angelman syndrome and ASD.


Subject(s)
Nerve Tissue Proteins/physiology , Neurogenesis/genetics , RNA, Messenger/physiology , Ubiquitin-Protein Ligases/genetics , 3' Untranslated Regions/genetics , Animals , Dendrites/ultrastructure , Excitatory Postsynaptic Potentials/physiology , Frameshift Mutation , HEK293 Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , MicroRNAs/genetics , Miniature Postsynaptic Potentials/physiology , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Neurons/ultrastructure , Protein Biosynthesis , Protein Isoforms/genetics , Protein Isoforms/physiology , RNA Interference , RNA Splicing , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Rats , Transfection
10.
EMBO J ; 33(19): 2231-46, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25097251

ABSTRACT

Neurons employ a set of homeostatic plasticity mechanisms to counterbalance altered levels of network activity. The molecular mechanisms underlying homeostatic plasticity in response to increased network excitability are still poorly understood. Here, we describe a sequential homeostatic synaptic depression mechanism in primary hippocampal neurons involving miRNA-dependent translational regulation. This mechanism consists of an initial phase of synapse elimination followed by a reinforcing phase of synaptic downscaling. The activity-regulated microRNA miR-134 is necessary for both synapse elimination and the structural rearrangements leading to synaptic downscaling. Results from miR-134 inhibition further uncover a differential requirement for GluA1/2 subunits for the functional expression of homeostatic synaptic depression. Downregulation of the miR-134 target Pumilio-2 in response to chronic activity, which selectively occurs in the synapto-dendritic compartment, is required for miR-134-mediated homeostatic synaptic depression. We further identified polo-like kinase 2 (Plk2) as a novel target of Pumilio-2 involved in the control of GluA2 surface expression. In summary, we have described a novel pathway of homeostatic plasticity that stabilizes neuronal circuits in response to increased network activity.


Subject(s)
Gene Expression Regulation , Hippocampus/metabolism , Homeostasis/physiology , MicroRNAs/genetics , Neurons/metabolism , RNA-Binding Proteins/metabolism , Synapses/physiology , Animals , Blotting, Western , Cells, Cultured , Electrophysiology , Fluorescent Antibody Technique , Hippocampus/embryology , Immunoprecipitation , Neuronal Plasticity , Neurons/cytology , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Cell Mol Life Sci ; 71(20): 3987-4005, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25008044

ABSTRACT

Dendritic mRNA transport and local translation in the postsynaptic compartment play an important role in synaptic plasticity, learning and memory. Local protein synthesis at the synapse has to be precisely orchestrated by a plethora of factors including RNA binding proteins as well as microRNAs, an extensive class of small non-coding RNAs. By binding to complementary sequences in target mRNAs, microRNAs fine-tune protein synthesis and thereby represent critical regulators of gene expression at the post-transcriptional level. Research over the last years identified an entire network of dendritic microRNAs that fulfills an essential role in synapse development and physiology. Recent studies provide evidence that these small regulatory molecules are highly regulated themselves, at the level of expression as well as function. The importance of microRNAs for correct function of the nervous system is reflected by an increasing number of studies linking dysregulation of microRNA pathways to neurological disorders. By focusing on three extensively studied examples (miR-132, miR-134, miR-138), this review will attempt to illustrate the complex regulatory roles of dendritic microRNAs at the synapse and their implications for pathological conditions.


Subject(s)
Dendrites/metabolism , MicroRNAs/metabolism , Animals , Dendrites/immunology , Humans , Nervous System/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Neuronal Plasticity , RNA-Binding Proteins/metabolism , Synapses/metabolism
12.
Genes Dev ; 27(9): 991-6, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23651854

ABSTRACT

Specific microRNAs (miRNAs), including miR-134, localize to neuronal dendrites, where they control synaptic protein synthesis and plasticity. However, the mechanism of miRNA transport is unknown. We found that the neuronal precursor-miRNA-134 (pre-miR-134) accumulates in dendrites of hippocampal neurons and at synapses in vivo. Dendritic localization of pre-miR-134 is mediated by the DEAH-box helicase DHX36, which directly associates with the pre-miR-134 terminal loop. DHX36 function is required for miR-134-dependent inhibition of target gene expression and the control of dendritic spine size. Dendritically localized pre-miR-134 could provide a local source of miR-134 that can be mobilized in an activity-dependent manner during plasticity.


Subject(s)
DEAD-box RNA Helicases/metabolism , Dendrites/enzymology , MicroRNAs/metabolism , Animals , Cells, Cultured , DEAD-box RNA Helicases/genetics , Dendritic Spines/metabolism , Gene Expression Regulation, Developmental , Hippocampus/metabolism , Rats , Synaptosomes/metabolism
14.
Cell ; 141(4): 618-31, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20478254

ABSTRACT

Adaptation to different levels of illumination is central to the function of the retina. Here, we demonstrate that levels of the miR-183/96/182 cluster, miR-204, and miR-211 are regulated by different light levels in the mouse retina. Concentrations of these microRNAs were downregulated during dark adaptation and upregulated in light-adapted retinas, with rapid decay and increased transcription being responsible for the respective changes. We identified the voltage-dependent glutamate transporter Slc1a1 as one of the miR-183/96/182 targets in photoreceptor cells. We found that microRNAs in retinal neurons decay much faster than microRNAs in nonneuronal cells. The high turnover is also characteristic of microRNAs in hippocampal and cortical neurons, and neurons differentiated from ES cells in vitro. Blocking activity reduced turnover of microRNAs in neuronal cells while stimulation with glutamate accelerated it. Our results demonstrate that microRNA metabolism in neurons is higher than in most other cells types and linked to neuronal activity.


Subject(s)
MicroRNAs/metabolism , Neurons/metabolism , Animals , Dark Adaptation , Down-Regulation , Embryonic Stem Cells , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Mice , Photoreceptor Cells, Vertebrate/metabolism , Retinal Neurons/metabolism , Up-Regulation
15.
Nat Cell Biol ; 11(6): 705-16, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19465924

ABSTRACT

The microRNA pathway has been implicated in the regulation of synaptic protein synthesis and ultimately in dendritic spine morphogenesis, a phenomenon associated with long-lasting forms of memory. However, the particular microRNAs (miRNAs) involved are largely unknown. Here we identify specific miRNAs that function at synapses to control dendritic spine structure by performing a functional screen. One of the identified miRNAs, miR-138, is highly enriched in the brain, localized within dendrites and negatively regulates the size of dendritic spines in rat hippocampal neurons. miR-138 controls the expression of acyl protein thioesterase 1 (APT1), an enzyme regulating the palmitoylation status of proteins that are known to function at the synapse, including the alpha(13) subunits of G proteins (Galpha(13)). RNA-interference-mediated knockdown of APT1 and the expression of membrane-localized Galpha(13) both suppress spine enlargement caused by inhibition of miR-138, suggesting that APT1-regulated depalmitoylation of Galpha(13) might be an important downstream event of miR-138 function. Our results uncover a previously unknown miRNA-dependent mechanism in neurons and demonstrate a previously unrecognized complexity of miRNA-dependent control of dendritic spine morphogenesis.


Subject(s)
Dendritic Spines , MicroRNAs/metabolism , Synapses , Thiolester Hydrolases/metabolism , Animals , Base Sequence , Cell Line , Dendritic Spines/enzymology , Dendritic Spines/ultrastructure , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Gene Expression Profiling , Hippocampus/cytology , Humans , Lipoylation , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Molecular Sequence Data , Morphogenesis , Neurons/cytology , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Rats , Receptors, Glutamate/metabolism , Synapses/metabolism , Synapses/ultrastructure , Thiolester Hydrolases/antagonists & inhibitors , Thiolester Hydrolases/genetics
16.
J Cell Mol Med ; 12(5A): 1466-76, 2008.
Article in English | MEDLINE | ID: mdl-18624757

ABSTRACT

The development and function of neuronal circuits within the brain are orchestrated by sophisticated gene regulatory mechanisms. Recently, microRNAs have emerged as a novel class of small RNAs that fine-tune protein synthesis. microRNAs are abundantly expressed in the vertebrate nervous system, where they contribute to the specification of neuronal cell identity. Moreover, microRNAs also play an important role in mature neurons. This review summarizes the current knowledge about the function of microRNAs in the nervous system with special emphasis on synapse formation and plasticity. The second part of this work will discuss the potential involvement of microRNAs in neurologic diseases. The study of brain microRNAs promises to expand our understanding of the mechanisms underlying higher cognitive functions and neurologic diseases.


Subject(s)
Gene Expression Regulation/genetics , MicroRNAs/genetics , Synapses/metabolism , Animals , Biological Transport , Dendrites/genetics , Disease , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...