Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
1.
Adv Sci (Weinh) ; 11(17): e2306630, 2024 May.
Article in English | MEDLINE | ID: mdl-38493494

ABSTRACT

The modification of synaptic and neural connections in adults, including the formation and removal of synapses, depends on activity-dependent synaptic and structural plasticity. MicroRNAs (miRNAs) play crucial roles in regulating these changes by targeting specific genes and regulating their expression. The fact that somatic and dendritic activity in neurons often occurs asynchronously highlights the need for spatial and dynamic regulation of protein synthesis in specific milieu and cellular loci. MicroRNAs, which can show distinct patterns of enrichment, help to establish the localized distribution of plasticity-related proteins. The recent study using atomic force microscopy (AFM)-based nanoscale imaging reveals that the abundance of miRNA(miR)-134 is inversely correlated with the functional activity of dendritic spine structures. However, the miRNAs that are selectively upregulated in potentiated synapses, and which can thereby support prospective changes in synaptic efficacy, remain largely unknown. Using AFM force imaging, significant increases in miR-132 in the dendritic regions abutting functionally-active spines is discovered. This study provides evidence for miR-132 as a novel positive miRNA regulator residing in dendritic shafts, and also suggests that activity-dependent miRNAs localized in distinct sub-compartments of neurons play bi-directional roles in controlling synaptic transmission and synaptic plasticity.


Subject(s)
MicroRNAs , Microscopy, Atomic Force , Neuronal Plasticity , Synapses , Animals , Mice , Dendritic Spines/metabolism , Dendritic Spines/genetics , Dendritic Spines/ultrastructure , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Microscopy, Atomic Force/methods , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Neurons/metabolism , Synapses/metabolism , Synapses/genetics
2.
Int J Mol Sci ; 24(2)2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36675068

ABSTRACT

Stress is a key risk factor in the onset of neuropsychiatric disorders. The study of the mechanisms underlying stress response is important to understand the etiopathogenetic mechanisms and identify new putative therapeutic targets. In this context, microRNAs (miRNAs) have emerged as key regulators of the complex patterns of gene/protein expression changes in the brain, where they have a crucial role in the regulation of neuroplasticity, neurogenesis, and neuronal differentiation. Among them, miR-135a-5p has been associated with stress response, synaptic plasticity, and the antidepressant effect in different brain areas. Here, we used acute unavoidable foot-shock stress (FS) and chronic mild stress (CMS) on male rats to study whether miR-135a-5p was involved in stress-induced changes in the prefrontal cortex (PFC). Both acute and chronic stress decreased miR-135a-5p levels in the PFC, although after CMS the reduction was induced only in animals vulnerable to CMS, according to a sucrose preference test. MiR-135a-5p downregulation in the primary neurons reduced dendritic spine density, while its overexpression exerted the opposite effect. Two bioinformatically predicted target genes, Kif5c and Cplx1/2, were increased in FS rats 24 h after stress. Altogether, we found that miR-135a-5p might play a role in stress response in PFC involving synaptic mechanisms.


Subject(s)
MicroRNAs , Prefrontal Cortex , Stress, Physiological , Stress, Psychological , Animals , Male , Rats , Down-Regulation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Neurons/metabolism , Neurons/pathology , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Prefrontal Cortex/physiology , Acute Disease/psychology , Chronic Disease/psychology , Stress, Physiological/genetics , Stress, Psychological/genetics , Stress, Psychological/psychology , Synapses/genetics , Synapses/metabolism , Synapses/pathology , Dendritic Spines/genetics , Dendritic Spines/metabolism , Dendritic Spines/pathology
3.
Stress ; 26(1): 1-14, 2023 01.
Article in English | MEDLINE | ID: mdl-36520154

ABSTRACT

Bombesin receptor-activated protein (BRAP) and its homologous protein in mice, which is encoded by bc004004 gene, were expressed abundantly in brain tissues with unknown functions. We treated bc004004-/- mice with chronic unpredictable mild stress (CUMS) to test whether those mice were more vulnerable to stress-related disorders. The results of forced swimming test, sucrose preference test, and open field test showed that after being treated with CUMS for 28 days or 35 days both bc004004-/- and bc004004+/+ mice exhibited behavioural changes and there was no significant difference between bc004004+/+ and bc004004-/-. However, behavioural changes were observed only in bc004004-/- mice after being exposed to CUMS for 21 days, but not in bc004004+/+ after 21-day CUMS exposure, indicating that lack of BRAP homologous protein may cause vulnerability to stress-related disorders in mice. In addition, bc004004-/- mice showed a reduction in recognition memory as revealed by novel object recognition test. Since memory changes and stress related behavioural changes are all closely related to the hippocampus function we further analyzed the changes of dendrites and synapses of hippocampal neurons as well as expression levels of some proteins closely related to synaptic function. bc004004-/- mice exhibited decreased dendritic lengths and increased amount of immature spines, as well as altered expression pattern of synaptic related proteins including GluN2A, synaptophysin and BDNF in the hippocampus. Those findings suggest that BRAP homologous protein may have a protective effect on the behavioural response to stress via regulating dendritic spine formation and synaptic plasticity in the hippocampus.


Subject(s)
Bombesin , Dendritic Spines , Hippocampus , Neuronal Plasticity , Receptors, Bombesin , Stress, Psychological , Animals , Mice , Bombesin/genetics , Bombesin/metabolism , Chronic Disease , Dendritic Spines/genetics , Dendritic Spines/metabolism , Dendritic Spines/pathology , Depression/genetics , Depression/metabolism , Depression/pathology , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Receptors, Bombesin/genetics , Receptors, Bombesin/metabolism , Stress, Psychological/genetics , Stress, Psychological/metabolism , Stress, Psychological/pathology
4.
FEBS J ; 289(8): 2282-2300, 2022 04.
Article in English | MEDLINE | ID: mdl-33511762

ABSTRACT

Autism spectrum disorder (ASD) is increasingly recognized as a condition of altered brain connectivity. As synapses are fundamental subcellular structures for neuronal connectivity, synaptic pathophysiology has become one of central themes in autism research. Reports disagree upon whether the density of dendritic spines, namely excitatory synapses, is increased or decreased in ASD and whether the protein synthesis that is critical for dendritic spine formation and function is upregulated or downregulated. Here, we review recent evidence supporting a subgroup of ASD models with decreased dendritic spine density (hereafter ASD-DSD), including Nf1 and Vcp mutant mice. We discuss the relevance of branched-chain amino acid (BCAA) insufficiency in relation to unmet protein synthesis demand in ASD-DSD. In contrast to ASD-DSD, ASD models with hyperactive mammalian target of rapamycin (mTOR) may represent the opposite end of the disease spectrum, often characterized by increases in protein synthesis and dendritic spine density (denoted ASD-ISD). Finally, we propose personalized dietary leucine as a strategy tailored to balancing protein synthesis demand, thereby ameliorating dendritic spine pathophysiologies and autism-related phenotypes in susceptible patients, especially those with ASD-DSD.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autistic Disorder/genetics , Autistic Disorder/metabolism , Dendritic Spines/genetics , Dendritic Spines/metabolism , Humans , Mammals , Mice , Neurons/metabolism , Synapses/metabolism
5.
Cells ; 10(12)2021 12 07.
Article in English | MEDLINE | ID: mdl-34943950

ABSTRACT

In day-to-day life, we often choose between pursuing familiar behaviors that have been rewarded in the past or adjusting behaviors when new strategies might be more fruitful. The dorsomedial striatum (DMS) is indispensable for flexibly arbitrating between old and new behavioral strategies. The way in which DMS neurons host stable connections necessary for sustained flexibility is still being defined. An entry point to addressing this question may be the structural scaffolds on DMS neurons that house synaptic connections. We find that the non-receptor tyrosine kinase Proline-rich tyrosine kinase 2 (Pyk2) stabilizes both dendrites and spines on striatal medium spiny neurons, such that Pyk2 loss causes dendrite arbor and spine loss. Viral-mediated Pyk2 silencing in the DMS obstructs the ability of mice to arbitrate between rewarded and non-rewarded behaviors. Meanwhile, the overexpression of Pyk2 or the closely related focal adhesion kinase (FAK) enhances this ability. Finally, experiments using combinatorial viral vector strategies suggest that flexible, Pyk2-dependent action involves inputs from the medial prefrontal cortex (mPFC), but not the ventrolateral orbitofrontal cortex (OFC). Thus, Pyk2 stabilizes the striatal medium spiny neuron structure, likely providing substrates for inputs, and supports the capacity of mice to arbitrate between novel and familiar behaviors, including via interactions with the medial-prefrontal cortex.


Subject(s)
Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 2/genetics , Neurons/metabolism , Prefrontal Cortex/metabolism , Animals , Corpus Striatum/metabolism , Dendrites/genetics , Dendrites/metabolism , Dendritic Spines/genetics , Dendritic Spines/metabolism , Humans , Mice , Neostriatum/metabolism , Neurons/pathology , Synaptic Transmission/genetics
6.
Biochem Biophys Res Commun ; 584: 107-115, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34781202

ABSTRACT

Dendritic spines are the postsynaptic structure to mediate signal transduction in neural circuitry, whose function and plasticity are regulated by organization of their molecular architecture and by the expression of target genes and proteins. EphB2, a member of the Eph receptor tyrosine kinase family, potentiates dendritic spine maturation through cytoskeleton reorganization and protein trafficking. However, the transcriptional mechanisms underlying prolonged activation of EphB2 signaling during dendritic spine morphogenesis are unknown. Herein, we performed transcriptional profiling by stimulating EphB2 signaling and identified differentially expressed genes implicated in pivotal roles at synapses. Notably, we characterized an F-actin binding protein, Annexin A1, whose expression was induced by EphB2 signaling; the promotor activity of its coding gene Anxa1 is regulated by the activity of CREB (cAMP-response element-binding protein). Knockdown of Annexin A1 led to a significant reduction of mature dendritic spines without an obvious deficit in the complexity of dendrites. Altogether, our findings suggest that EphB2-induced, CREB-dependent Annexin A1 expression plays a key role in regulating dendritic spine morphology.


Subject(s)
Annexin A1/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Dendritic Spines/genetics , Receptor, EphB2/genetics , Annexin A1/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Dendritic Spines/physiology , Gene Expression Profiling/methods , Gene Ontology , Gene Regulatory Networks/genetics , HEK293 Cells , Humans , Microscopy, Confocal , Morphogenesis/genetics , Neurons/metabolism , Protein Interaction Maps/genetics , RNA-Seq/methods , Receptor, EphB2/metabolism , Signal Transduction/genetics , Synapses/genetics , Synapses/physiology
7.
Nat Commun ; 12(1): 6444, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34750364

ABSTRACT

Synaptic pruning during adolescence is important for appropriate neurodevelopment and synaptic plasticity. Aberrant synaptic pruning may underlie a variety of brain disorders such as schizophrenia, autism and anxiety. Dopamine D2 receptor (Drd2) is associated with several neuropsychiatric diseases and is the target of some antipsychotic drugs. Here we generate self-reporting Drd2 heterozygous (SR-Drd2+/-) rats to simultaneously visualize Drd2-positive neurons and downregulate Drd2 expression. Time course studies on the developing anterior cingulate cortex (ACC) from control and SR-Drd2+/- rats reveal important roles of Drd2 in regulating synaptic pruning rather than synapse formation. Drd2 also regulates LTD, a form of synaptic plasticity which includes some similar cellular/biochemical processes as synaptic pruning. We further demonstrate that Drd2 regulates synaptic pruning via cell-autonomous mechanisms involving activation of mTOR signaling. Deficits of Drd2-mediated synaptic pruning in the ACC during adolescence lead to hyper-glutamatergic function and anxiety-like behaviors in adulthood. Taken together, our results demonstrate important roles of Drd2 in cortical synaptic pruning.


Subject(s)
Gyrus Cinguli/physiology , Neuronal Plasticity/physiology , Receptors, Dopamine D2/physiology , Signal Transduction/physiology , Animals , Animals, Genetically Modified , Dendritic Spines/genetics , Dendritic Spines/physiology , Gene Knockout Techniques , Gyrus Cinguli/cytology , Gyrus Cinguli/metabolism , Heterozygote , Inhibitory Postsynaptic Potentials/genetics , Inhibitory Postsynaptic Potentials/physiology , Mutation , Neuronal Plasticity/genetics , Neurons/cytology , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques/methods , Rats, Sprague-Dawley , Receptors, Dopamine D2/genetics , Signal Transduction/genetics , Synapses/genetics , Synapses/physiology , Time Factors
8.
J Neurosci ; 41(37): 7768-7778, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34353896

ABSTRACT

We recently identified an autism spectrum disorder/intellectual disability (ASD/ID)-related de novo mutation hotspot in the Rac1-activating GEF1 domain of the protein Trio. Trio is a Rho guanine nucleotide exchange factor (RhoGEF) that is essential for glutamatergic synapse function. An ASD/ID-related mutation identified in Trio's GEF1 domain, Trio D1368V, produces a pathologic increase in glutamatergic synaptogenesis, suggesting that Trio is coupled to synaptic regulatory mechanisms that govern glutamatergic synapse formation. However, the molecular mechanisms by which Trio regulates glutamatergic synapses are largely unexplored. Here, using biochemical methods, we identify an interaction between Trio and the synaptogenic protein Neuroligin 1 (NLGN1) in the brain. Molecular biological approaches were then combined with super-resolution dendritic spine imaging and whole-cell voltage-clamp electrophysiology in hippocampal slices from male and female rats to examine the impact ASD/ID-related Trio mutations have on NLGN1-mediated synaptogenesis. We find that an ASD/ID-related mutation in Trio's eighth spectrin repeat region, Trio N1080I, inhibits Trio's interaction with NLGN1 and prevents Trio D1368V-mediated synaptogenesis. Inhibiting Trio's interaction with NLGN1 via Trio N1080I blocked NLGN1-mediated synaptogenesis and increases in synaptic NMDA Receptor function but not NLGN1-mediated increases in synaptic AMPA Receptor function. Finally, we show that the aberrant synaptogenesis produced by Trio D1368V is dependent on NLGN signaling. Our findings demonstrate that ASD/ID-related mutations in Trio are able to pathologically increase as well as decrease NLGN-mediated effects on glutamatergic neurotransmission, and point to an NLGN1-Trio interaction as part of a key pathway involved in ASD/ID etiology.SIGNIFICANCE STATEMENT A number of genes have been implicated in the development of autism spectrum disorder/intellectual disability (ASD/ID) in humans. It is now important to identify relationships between these genes to uncover specific cellular regulatory pathways that contribute to these disorders. In this study, we discover that two glutamatergic synapse regulatory proteins implicated in ASD/ID, Trio and Neuroligin 1, interact with one another to promote glutamatergic synaptogenesis. We also identify ASD/ID-related mutations in Trio that either inhibit or augment Neuroligin 1-mediated glutamatergic synapse formation. Together, our results identify a synaptic regulatory pathway that, when disrupted, likely contributes to the development of ASD/ID. Going forward, it will be important to determine whether this pathway represents a point of convergence of other proteins implicated in ASD/ID.


Subject(s)
Autism Spectrum Disorder/genetics , Cell Adhesion Molecules, Neuronal/genetics , Intellectual Disability/genetics , Mutation , Synapses/genetics , Animals , Autism Spectrum Disorder/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Dendritic Spines/genetics , Dendritic Spines/metabolism , Excitatory Postsynaptic Potentials/physiology , Female , Hippocampus/metabolism , Intellectual Disability/metabolism , Male , Neurogenesis/physiology , Rats , Rats, Sprague-Dawley , Synapses/metabolism
9.
Int J Mol Sci ; 22(11)2021 Jun 05.
Article in English | MEDLINE | ID: mdl-34198910

ABSTRACT

Changes in structural and functional neuroplasticity have been implicated in various neurological disorders. Sterol regulatory element-binding protein (SREBP)-1c is a critical regulatory molecule of lipid homeostasis in the brain. Recently, our findings have shown the potential involvement of SREBP-1c deficiency in the alteration of novel modulatory molecules in the hippocampus and occurrence of schizophrenia-like behaviors in mice. However, the possible underlying mechanisms, related to neuronal plasticity in the hippocampus, are yet to be elucidated. In this study, we investigated the hippocampus-dependent memory function and neuronal architecture of hippocampal neurons in SREBP-1c knockout (KO) mice. During the passive avoidance test, SREBP-1c KO mice showed memory impairment. Based on Golgi staining, the dendritic complexity, length, and branch points were significantly decreased in the apical cornu ammonis (CA) 1, CA3, and dentate gyrus (DG) subregions of the hippocampi of SREBP-1c KO mice, compared with those of wild-type (WT) mice. Additionally, significant decreases in the dendritic diameters were detected in the CA3 and DG subregions, and spine density was also significantly decreased in the apical CA3 subregion of the hippocampi of KO mice, compared with that of WT mice. Alterations in the proportions of stubby and thin-shaped dendritic spines were observed in the apical subcompartments of CA1 and CA3 in the hippocampi of KO mice. Furthermore, the corresponding differential decreases in the levels of SREBP-1 expression in the hippocampal subregions (particularly, a significant decrease in the level in the CA3) were detected by immunofluorescence. This study suggests that the contributions of SREBP-1c to the structural plasticity of the mouse hippocampus may have underlain the behavioral alterations. These findings offer insights into the critical role of SREBP-1c in hippocampal functioning in mice.


Subject(s)
Dendritic Spines/genetics , Memory/physiology , Neurons/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Animals , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Dendritic Spines/pathology , Gene Expression Regulation/genetics , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Knockout , Neuronal Plasticity/genetics , Neurons/pathology , Sterol Regulatory Element Binding Protein 1/deficiency
10.
Neuroreport ; 32(10): 833-839, 2021 07 07.
Article in English | MEDLINE | ID: mdl-34029289

ABSTRACT

ATP-binding cassettes C1 (ABCC1s) are expressed in the neurons of the brain, but their function in neurological diseases is far from clear. In this study, we investigated the role of ABCC1 in the hippocampus in cocaine-associated memory and spine plasticity. We also investigated the role of ABCC1 in AMPA receptors (AMPARs) surface expression in primary prefrontal cortex (PFC) neurons following dopamine treatment, which was used to mimic exposure to cocaine. We found that cocaine increased ABCC1 expression in the hippocampus, and ABCC1-siRNA blocked cocaine-induced place preference. Furthermore, a morphological study showed that ABCC1-siRNA reduced the total spine density, including thin, stubby and mushroom spines in both cocaine and basal treatments compared with controls. Meanwhile, in vitro tests showed that ABCC1-siRNA decreased GluA1 and GluA2 surface expression induced by dopamine, while a decreased number of synapses in primary PFC neurons was observed following dopamine treatment. The data show that ABCC1 in the hippocampus is critically involved in cocaine-associated memory and spine plasticity and that dopamine induces AMPARs surface expression in primary PFC neurons. ABCC1 is thus presented as a new signaling molecule involved in cocaine addiction, which may provide a new target for the treatment of cocaine addiction.


Subject(s)
Cocaine/administration & dosage , Memory/drug effects , Multidrug Resistance-Associated Proteins/biosynthesis , Neuronal Plasticity/drug effects , Receptors, AMPA/biosynthesis , Animals , Animals, Newborn , Cells, Cultured , Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/metabolism , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Dendritic Spines/drug effects , Dendritic Spines/genetics , Dendritic Spines/metabolism , Dopamine Uptake Inhibitors/administration & dosage , Gene Expression , Male , Memory/physiology , Mice , Mice, Inbred C57BL , Multidrug Resistance-Associated Proteins/genetics , Neuronal Plasticity/physiology , Receptors, AMPA/genetics
11.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Article in English | MEDLINE | ID: mdl-33906942

ABSTRACT

Loss of the fragile X mental retardation protein (FMRP) causes fragile X syndrome (FXS). FMRP is widely thought to repress protein synthesis, but its translational targets and modes of control remain in dispute. We previously showed that genetic removal of p70 S6 kinase 1 (S6K1) corrects altered protein synthesis as well as synaptic and behavioral phenotypes in FXS mice. In this study, we examined the gene specificity of altered messenger RNA (mRNA) translation in FXS and the mechanism of rescue with genetic reduction of S6K1 by carrying out ribosome profiling and RNA sequencing on cortical lysates from wild-type, FXS, S6K1 knockout, and double knockout mice. We observed reduced ribosome footprint (RF) abundance in the majority of differentially translated genes in the cortices of FXS mice. We used molecular assays to discover evidence that the reduction in RF abundance reflects an increased rate of ribosome translocation, which is captured as a decrease in the number of translating ribosomes at steady state and is normalized by inhibition of S6K1. We also found that genetic removal of S6K1 prevented a positive-to-negative gradation of alterations in translation efficiencies (RF/mRNA) with coding sequence length across mRNAs in FXS mouse cortices. Our findings reveal the identities of dysregulated mRNAs and a molecular mechanism by which reduction of S6K1 prevents altered translation in FXS.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Protein Biosynthesis , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Animals , Dendritic Spines/genetics , Dendritic Spines/pathology , Disease Models, Animal , Fragile X Syndrome/pathology , Fragile X Syndrome/therapy , Humans , Mice , Mice, Knockout , Mutation/genetics , Neurons/metabolism , Neurons/pathology , Open Reading Frames/genetics , RNA, Messenger/genetics
12.
J Neurosci ; 41(14): 3068-3081, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33622779

ABSTRACT

Dendritic spines act as the receptive contacts at most excitatory synapses. Spines are enriched in a network of actin filaments comprised of two kinetically distinct pools. The majority of spine actin is highly dynamic and regulates spine size, structural plasticity, and postsynaptic density organization. The remainder of the spine actin network is more stable, but the function of this minor actin population is not well understood, as tools to study it have not been available. Previous work has shown that disruption of the Abl2/Arg nonreceptor tyrosine kinase in mice compromises spine stability and size. Here, using cultured hippocampal neurons pooled from both sexes of mice, we provide evidence that binding to cortactin tethers Abl2 in spines, where Abl2 and cortactin maintain the small pool of stable actin required for dendritic spine stability. Using fluorescence recovery after photobleaching of GFP-actin, we find that disruption of Abl2:cortactin interactions eliminates stable actin filaments in dendritic spines, significantly reducing spine density. A subset of spines remaining after Abl2 depletion retain their stable actin pool and undergo activity-dependent spine enlargement, associated with increased cortactin and GluN2B levels. Finally, tonic increases in synaptic activity rescue spine loss following Abl2 depletion by promoting cortactin enrichment in vulnerable spines. Together, our findings strongly suggest that Abl2:cortactin interactions promote spine stability by maintaining pools of stable actin filaments in spines.SIGNIFICANCE STATEMENT Dendritic spines contain two kinetically distinct pools of actin. The more abundant, highly dynamic pool regulates spine shape, size, and plasticity. The function of the smaller, stable actin network is not well understood, as tools to study it have not been available. We demonstrate here that Abl2 and its substrate and interaction partner, cortactin, are essential to maintain the stable pool in spines. Depletion of the stable actin pool via disruption of Abl2 or cortactin, or interactions between the proteins, significantly reduces spine stability. We also provide evidence that tonic increases in synaptic activity promote spine stability via enrichment of cortactin in spines, suggesting that synaptic activity acts on the stable actin pool to stabilize dendritic spines.


Subject(s)
Actin Cytoskeleton/metabolism , Cortactin/metabolism , Dendritic Spines/metabolism , Protein-Tyrosine Kinases/metabolism , Actin Cytoskeleton/genetics , Actins/genetics , Actins/metabolism , Animals , Animals, Newborn , Cortactin/genetics , Dendritic Spines/genetics , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Protein Binding/physiology , Protein-Tyrosine Kinases/genetics
13.
Acta Pharmacol Sin ; 42(5): 679-690, 2021 May.
Article in English | MEDLINE | ID: mdl-33526871

ABSTRACT

Over the last decade, the roles of ß-arrestins in the treatment of neuropsychological diseases have become increasingly appreciated. Fluoxetine is the first selective serotonin reuptake inhibitor developed and is approved for the clinical treatment of depression. Emerging evidence suggests that fluoxetine can directly combine with the 5-HT receptor, which is a member of the G protein-coupled receptor (GPCR) family, in addition to suppressing the serotonin transporter. In this study, we prepared a chronic mild stress (CMS)-induced depression model with ß-arrestin2-/- mice and cultured adult neural stem cells (ANSCs) to investigate the involvement of the 5-HT receptor-ß-arrestin axis in the pathogenesis of depression and in the therapeutic effect of fluoxetine. We found that ß-arrestin2 deletion abolished the fluoxetine-mediated improvement in depression-like behaviors and monoamine neurotransmitter levels, although ß-arrestin2 knockout did not aggravate CMS-induced changes in mouse behaviors and neurotransmitters. Notably, the ß-arrestin2-/- mice had a shortened dendritic length and reduced dendritic spine density, as well as decreased neural precursor cells, compared to the WT mice under both basal and CMS conditions. We further found that ß-arrestin2 knockout decreased the number of proliferating cells in the hippocampal dentate gyrus and suppressed the proliferative capability of ANSCs in vitro. Moreover, ß-arrestin2 knockout aggravated the impairment of cell proliferation induced by corticosterone and further blocked the fluoxetine-mediated promotion of mouse hippocampal neurogenesis. Mechanistically, we found that the 5-HT2BR-ß-arrestin2-PI3K/Akt axis is essential to maintain the modulation of hippocampal neurogenesis in depressed mice. Our study may provide a promising target for the development of new antidepressant drugs.


Subject(s)
Antidepressive Agents/therapeutic use , Dentate Gyrus/drug effects , Depression/drug therapy , Fluoxetine/therapeutic use , Selective Serotonin Reuptake Inhibitors/therapeutic use , beta-Arrestin 2/metabolism , Animals , Cell Proliferation/drug effects , Cell Proliferation/genetics , Dendritic Spines/genetics , Dentate Gyrus/metabolism , Depression/metabolism , Gene Knockout Techniques , Male , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurotransmitter Agents/metabolism , Signal Transduction/drug effects , beta-Arrestin 2/genetics
14.
J Comp Neurol ; 529(6): 1099-1120, 2021 04 15.
Article in English | MEDLINE | ID: mdl-32785985

ABSTRACT

Lafora disease (LD) is a genetic and fatal form of neurodegenerative disorder characterized by myoclonic epilepsy and cognitive deficits. LD is caused by loss-of-function mutations in the EPM2A or the NHLRC1 gene. A major hallmark of LD is the presence of abnormal glycogen aggregates in neurons and other tissues. Functional studies on the genes have, therefore, mostly focused on glycogen metabolism. The physiological basis of cognitive deficits in LD is thus largely unexplored. Alterations in dendritic spine morphology are known in neurodevelopmental and neuropsychiatric disorders. We, therefore, analyzed the dendritic spine morphologies in pyramidal neurons of the hippocampal and Cortical layer V of the Epm2a or Nhlrc1 knockout mice brain. We found a significant increase in the density, length, and reduction in the width of the dendritic spines in Postnatal day 21 to 12-month-old LD animals. Similar observations were made in the primary cultures of neurons derived from the hippocampi of the embryonic brain, suggesting that the aberrant spine phenotype could be a developmental defect in LD. We also looked at the cognitive and behavioral deficits as a possible readout of the spine abnormalities. The LD animals exhibited hyperactivity, reduced anxiety-like behavior, and deficits in the spatial and nonspatial memory. Such abnormalities were seen in the younger (1-2 months) as well as the older (7-8 months) age groups. Taken together, our results suggest that the dendritic spine abnormalities are primary developmental defects in the LD model and these defects might underlie some of the symptoms, including cognitive deficits, in LD.


Subject(s)
Cerebral Cortex/pathology , Cognitive Dysfunction/pathology , Dendritic Spines/pathology , Hippocampus/pathology , Lafora Disease/pathology , Memory/physiology , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Dendritic Spines/genetics , Dendritic Spines/metabolism , Female , Hippocampus/metabolism , Lafora Disease/genetics , Lafora Disease/metabolism , Male , Mice , Mice, Knockout , Pregnancy , Protein Tyrosine Phosphatases, Non-Receptor/deficiency , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics
15.
Cereb Cortex ; 31(4): 2205-2219, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33251537

ABSTRACT

Changes in the shape and size of the dendritic spines are critical for synaptic transmission. These morphological changes depend on dynamic assembly of the actin cytoskeleton and occur differently in various types of neurons. However, how the actin dynamics are regulated in a neuronal cell type-specific manner remains largely unknown. We show that Fhod3, a member of the formin family proteins that mediate F-actin assembly, controls the dendritic spine morphogenesis of specific subpopulations of cerebrocortical pyramidal neurons. Fhod3 is expressed specifically in excitatory pyramidal neurons within layers II/III and V of restricted areas of the mouse cerebral cortex. Immunohistochemical and biochemical analyses revealed the accumulation of Fhod3 in postsynaptic spines. Although targeted deletion of Fhod3 in the brain did not lead to any defects in the gross or histological appearance of the brain, the dendritic spines in pyramidal neurons within presumptive Fhod3-positive areas were morphologically abnormal. In primary cultures prepared from the Fhod3-depleted cortex, defects in spine morphology were only detected in Fhod3 promoter-active cells, a small population of pyramidal neurons, and not in Fhod3 promoter-negative pyramidal neurons. Thus, Fhod3 plays a crucial role in dendritic spine morphogenesis only in a specific population of pyramidal neurons in a cell type-specific manner.


Subject(s)
Cerebral Cortex/metabolism , Dendritic Spines/metabolism , Formins/biosynthesis , Pyramidal Cells/metabolism , Animals , Cells, Cultured , Cerebral Cortex/ultrastructure , Dendritic Spines/genetics , Dendritic Spines/ultrastructure , Formins/genetics , HEK293 Cells , Humans , Mice , Mice, Transgenic , Pyramidal Cells/ultrastructure
16.
Neurobiol Learn Mem ; 178: 107364, 2021 02.
Article in English | MEDLINE | ID: mdl-33340671

ABSTRACT

PCDH10 is a gene associated with Autism Spectrum Disorder. It is involved in the growth of thalamocortical projections and dendritic spine elimination. Previously, we characterized Pcdh10 haploinsufficient mice (Pcdh10+/- mice) and found male-specific social deficits and dark phase hypoactivity. Pcdh10+/- males exhibit increased dendritic spine density of immature morphology, decreased NMDAR expression, and decreased gamma synchronization in the basolateral amygdala (BLA). Here, we further characterize Pcdh10+/- mice by testing for fear memory, which relies on BLA function. We used both male and female Pcdh10+/- mice and their wild-type littermates at two ages, juvenile and adult, and in two learning paradigms, cued and contextual fear conditioning. We found that males at both ages and in both assays exhibited fear conditioning deficits, but females were only impaired as adults in the cued condition. These data are further evidence for male-specific alterations in BLA-related behaviors in Pcdh10+/- mice and suggest that these mice may be a useful model for dissecting male specific brain and behavioral phenotypes relevant to social and emotional behaviors.


Subject(s)
Basolateral Nuclear Complex/physiopathology , Cadherins/genetics , Conditioning, Classical/physiology , Fear/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Age Factors , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/physiopathology , Basolateral Nuclear Complex/metabolism , Cadherins/metabolism , Dendritic Spines/genetics , Dendritic Spines/metabolism , Female , Male , Mice , Mice, Knockout , Protocadherins , Receptors, N-Methyl-D-Aspartate/genetics , Sex Factors
17.
FASEB J ; 35(1): e21092, 2021 01.
Article in English | MEDLINE | ID: mdl-33378124

ABSTRACT

Myosin 18Aα is a myosin 2-like protein containing unique N- and C-terminal protein interaction domains that co-assembles with myosin 2. One protein known to bind to myosin 18Aα is ß-Pix, a guanine nucleotide exchange factor (GEF) for Rac1 and Cdc42 that has been shown to promote dendritic spine maturation by activating the assembly of actin and myosin filaments in spines. Here, we show that myosin 18A⍺ concentrates in the spines of cerebellar Purkinje neurons via co-assembly with myosin 2 and through an actin binding site in its N-terminal extension. miRNA-mediated knockdown of myosin 18A⍺ results in a significant defect in spine maturation that is rescued by an RNAi-immune version of myosin 18A⍺. Importantly, ß-Pix co-localizes with myosin 18A⍺ in spines, and its spine localization is lost upon myosin 18A⍺ knockdown or when its myosin 18A⍺ binding site is deleted. Finally, we show that the spines of myosin 18A⍺ knockdown Purkinje neurons contain significantly less F-actin and myosin 2. Together, these data argue that mixed filaments of myosin 2 and myosin 18A⍺ form a complex with ß-Pix in Purkinje neuron spines that promotes spine maturation by enhancing the assembly of actin and myosin filaments downstream of ß-Pix's GEF activity.


Subject(s)
Dendritic Spines/metabolism , Myosins/metabolism , Purkinje Cells/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Animals , Dendritic Spines/genetics , Gene Deletion , Mice , Myosin Type II/genetics , Myosin Type II/metabolism , Myosins/genetics , Rho Guanine Nucleotide Exchange Factors/genetics
18.
J Biomed Sci ; 27(1): 103, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-33256713

ABSTRACT

BACKGROUND: Dendritic spines, the actin-rich protrusions emerging from dendrites, are the subcellular locations of excitatory synapses in the mammalian brain. Many actin-regulating molecules modulate dendritic spine morphology. Since dendritic spines are neuron-specific structures, it is reasonable to speculate that neuron-specific or -predominant factors are involved in dendritic spine formation. KLHL17 (Kelch-like 17, also known as Actinfilin), an actin-binding protein, is predominantly expressed in brain. Human genetic study has indicated an association of KLHL17/Actinfilin with infantile spasms, a rare form of childhood epilepsy also resulting in autism and mental retardation, indicating that KLHL17/Actinfilin plays a role in neuronal function. However, it remains elusive if and how KLHL17/Actinfilin regulates neuronal development and brain function. METHODS: Fluorescent immunostaining and electrophysiological recording were performed to evaluate dendritic spine formation and activity in cultured hippocampal neurons. Knockdown and knockout of KLHL17/Actinfilin and expression of truncated fragments of KLHL17/Actinfilin were conducted to investigate the function of KLHL17/Actinfilin in neurons. Mouse behavioral assays were used to evaluate the role of KLHL17/Actinfilin in brain function. RESULTS: We found that KLHL17/Actinfilin tends to form circular puncta in dendritic spines and are surrounded by or adjacent to F-actin. Klhl17 deficiency impairs F-actin enrichment at dendritic spines. Knockdown and knockout of KLHL17/Actinfilin specifically impair dendritic spine enlargement, but not the density or length of dendritic spines. Both N-terminal Broad-Complex, Tramtrack and Bric-a-brac (BTB) domain and C-terminal Kelch domains of KLHL17/Actinfilin are required for F-actin remodeling and enrichment at dendritic spines, as well as dendritic spine enlargement. A reduction of postsynaptic and presynsptic markers at dendritic spines and altered mEPSC profiles due to Klhl17 deficiency evidence impaired synaptic activity in Klhl17-deficient neurons. Our behavioral assays further indicate that Klhl17 deficiency results in hyperactivity and reduced social interaction, strengthening evidence for the physiological role of KLHL17/Actinfilin. CONCLUSION: Our findings provide evidence that KLHL17/Actinfilin modulates F-actin remodeling and contributes to regulation of neuronal morphogenesis, maturation and activity, which is likely relevant to behavioral impairment in Klhl17-deficient mice. Trial registration Non-applicable.


Subject(s)
Autistic Disorder/genetics , Dendritic Spines/genetics , Microfilament Proteins/genetics , Spasms, Infantile/genetics , Animals , Dendritic Spines/metabolism , Disease Models, Animal , Female , Humans , Infant , Infant, Newborn , Male , Mice , Mice, Knockout , Microfilament Proteins/metabolism
19.
J Neurosci ; 40(50): 9552-9563, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33139402

ABSTRACT

Triggering receptor expressed on myeloid cells 2 (TREM2), a receptor exclusively expressed by microglia in the brain, modulates microglial immune homeostasis. Human genetic studies have shown that the loss-of-function mutations in TREM2 signaling are strongly associated with an elevated risk of age-related neurodegenerative diseases including Alzheimer's disease (AD). Numerous studies have investigated the impact of TREM2 deficiency in the pathogenic process of AD. However, the role of TREM2 in shaping neuronal and cognitive function during normal aging is underexplored. In the present study, we employed behavioral, electrophysiological, and biochemical approaches to assess cognitive and synaptic function in male and female young and aged TREM2-deficient (Trem2-/-) mice compared with age-matched, sex-matched, and genetic background-matched wild-type (WT) C57BL/6J controls. Young Trem2-/- mice exhibited normal cognitive function and synaptic plasticity but had increased dendritic spine density compared with young WT. Unexpectedly, aged Trem2-/- mice showed superior cognitive performance compared with aged WT controls. Consistent with the behavioral data, aged Trem2-/- mice displayed significantly enhanced hippocampal long-term potentiation (LTP) and increased dendritic spine density and synaptic markers compared with aged WT mice. Taken together, these findings suggest that loss of TREM2 affects the neuronal structure and confers resilience to age-related synaptic and cognitive impairment during non-pathogenic aging.SIGNIFICANCE STATEMENT Microglia are innate immune cells of the brain that orchestrates neurodevelopment, synaptic function, and immune response to environmental stimuli. Microglial triggering receptor expressed on myeloid cells 2 (TREM2) signaling plays pivotal roles in regulating these functions and loss of TREM2 signaling leads to increased risk of developing age-related neurologic disorders. However, the neurologic role of TREM2 in normal aging is poorly understood. The results of the present study unveil the positive impacts of TREM2 deficiency on cognitive and synaptic function during aging and suggest that TREM2 may exert detrimental effects on neuronal function. The possibility of age-related negative impacts from TREM2 is critically important since TREM2 has emerged as a major therapeutic target for Alzheimer's dementia.


Subject(s)
Aging/genetics , Cognition/physiology , Cognitive Dysfunction/genetics , Membrane Glycoproteins/genetics , Neuronal Plasticity/genetics , Receptors, Immunologic/genetics , Synapses/genetics , Aging/metabolism , Animals , Cognitive Dysfunction/metabolism , Dendritic Spines/genetics , Dendritic Spines/metabolism , Female , Hippocampus/metabolism , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Motor Activity/genetics , Neurons/metabolism , Receptors, Immunologic/metabolism , Synapses/metabolism
20.
Sci Rep ; 10(1): 16058, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32994505

ABSTRACT

Deletion of the autism candidate molecule neurobeachin (Nbea), a large PH-BEACH-domain containing neuronal protein, has been shown to affect synaptic function by interfering with neurotransmitter receptor targeting and dendritic spine formation. Previous analysis of mice lacking one allele of the Nbea gene identified impaired spatial learning and memory in addition to altered autism-related behaviours. However, no functional data from living heterozygous Nbea mice (Nbea+/-) are available to corroborate the behavioural phenotype. Here, we explored the consequences of Nbea haploinsufficiency on excitation/inhibition balance and synaptic plasticity in the intact hippocampal dentate gyrus of Nbea+/- animals in vivo by electrophysiological recordings. Based on field potential recordings, we show that Nbea+/- mice display enhanced LTP of the granule cell population spike, but no differences in basal synaptic transmission, synapse numbers, short-term plasticity, or network inhibition. These data indicate that Nbea haploinsufficiency causes remarkably specific alterations to granule cell excitability in vivo, which may contribute to the behavioural abnormalities in Nbea+/- mice and to related symptoms in patients.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Long-Term Potentiation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Animals , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Brain/metabolism , Dendritic Spines/genetics , Dendritic Spines/physiology , Dentate Gyrus/metabolism , Haploinsufficiency , Hippocampus/metabolism , Humans , Male , Membrane Proteins/metabolism , Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/genetics , Neurons/metabolism , Synapses/metabolism , Synaptic Transmission/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...