Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 177(5): 1346-1360.e24, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31080068

ABSTRACT

To decipher dynamic brain information processing, current genetically encoded calcium indicators (GECIs) are limited in single action potential (AP) detection speed, combinatorial spectral compatibility, and two-photon imaging depth. To address this, here, we rationally engineered a next-generation quadricolor GECI suite, XCaMPs. Single AP detection was achieved within 3-10 ms of spike onset, enabling measurements of fast-spike trains in parvalbumin (PV)-positive interneurons in the barrel cortex in vivo and recording three distinct (two inhibitory and one excitatory) ensembles during pre-motion activity in freely moving mice. In vivo paired recording of pre- and postsynaptic firing revealed spatiotemporal constraints of dendritic inhibition in layer 1 in vivo, between axons of somatostatin (SST)-positive interneurons and apical tufts dendrites of excitatory pyramidal neurons. Finally, non-invasive, subcortical imaging using red XCaMP-R uncovered somatosensation-evoked persistent activity in hippocampal CA1 neurons. Thus, the XCaMPs offer a critical enhancement of solution space in studies of complex neuronal circuit dynamics. VIDEO ABSTRACT.


Subject(s)
Action Potentials/physiology , Axons/metabolism , Cerebral Cortex/metabolism , Hippocampus/metabolism , Interneurons/metabolism , Pyramidal Cells/metabolism , Animals , Cerebral Cortex/cytology , Female , Hippocampus/cytology , Interneurons/cytology , Mice , Mice, Transgenic , Pyramidal Cells/cytology , Rats , Rats, Sprague-Dawley
2.
Cell ; 149(4): 886-98, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22579289

ABSTRACT

The Arc/Arg3.1 gene product is rapidly upregulated by strong synaptic activity and critically contributes to weakening synapses by promoting AMPA-R endocytosis. However, how activity-induced Arc is redistributed and determines the synapses to be weakened remains unclear. Here, we show targeting of Arc to inactive synapses via a high-affinity interaction with CaMKIIß that is not bound to calmodulin. Synaptic Arc accumulates in inactive synapses that previously experienced strong activation and correlates with removal of surface GluA1 from individual synapses. A lack of CaMKIIß either in vitro or in vivo resulted in loss of Arc upregulation in the silenced synapses. The discovery of Arc's role in "inverse" synaptic tagging that is specific for weaker synapses and prevents undesired enhancement of weak synapses in potentiated neurons reconciles essential roles of Arc both for the late phase of long-term plasticity and for reduction of surface AMPA-Rs in stimulated neurons.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cytoskeletal Proteins/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Synapses/metabolism , Animals , Cells, Cultured , Dendritic Spines/metabolism , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Rats, Sprague-Dawley
3.
J Neurosci ; 44(11)2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38286627

ABSTRACT

Dopamine neurons play crucial roles in pleasure, reward, memory, learning, and fine motor skills and their dysfunction is associated with various neuropsychiatric diseases. Dopamine receptors are the main target of treatment for neurologic and psychiatric disorders. Antipsychotics that antagonize the dopamine D2 receptor (DRD2) are used to alleviate the symptoms of these disorders but may also sometimes cause disabling side effects such as parkinsonism (catalepsy in rodents). Here we show that GPR143, a G-protein-coupled receptor for L-3,4-dihydroxyphenylalanine (L-DOPA), expressed in striatal cholinergic interneurons enhances the DRD2-mediated side effects of haloperidol, an antipsychotic agent. Haloperidol-induced catalepsy was attenuated in male Gpr143 gene-deficient (Gpr143-/y ) mice compared with wild-type (Wt) mice. Reducing the endogenous release of L-DOPA and preventing interactions between GPR143 and DRD2 suppressed the haloperidol-induced catalepsy in Wt mice but not Gpr143-/y mice. The phenotypic defect in Gpr143-/y mice was mimicked in cholinergic interneuron-specific Gpr143-/y (Chat-cre;Gpr143flox/y ) mice. Administration of haloperidol increased the phosphorylation of ribosomal protein S6 at Ser240/244 in the dorsolateral striatum of Wt mice but not Chat-cre;Gpr143flox/y mice. In Chinese hamster ovary cells stably expressing DRD2, co-expression of GPR143 increased cell surface expression level of DRD2, and L-DOPA application further enhanced the DRD2 surface expression. Shorter pauses in cholinergic interneuron firing activity were observed after intrastriatal stimulation in striatal slice preparations from Chat-cre;Gpr143flox/y mice compared with those from Wt mice. Together, these findings provide evidence that GPR143 regulates DRD2 function in cholinergic interneurons and may be involved in parkinsonism induced by antipsychotic drugs.


Subject(s)
Antipsychotic Agents , Parkinsonian Disorders , Receptors, Neurotransmitter , Humans , Mice , Male , Animals , Cricetinae , Haloperidol/pharmacology , Levodopa/adverse effects , Catalepsy/chemically induced , CHO Cells , Cricetulus , Antipsychotic Agents/adverse effects , Interneurons/metabolism , Cholinergic Agents/pharmacology , Eye Proteins/metabolism , Membrane Glycoproteins/metabolism
4.
Mol Psychiatry ; 28(2): 733-745, 2023 02.
Article in English | MEDLINE | ID: mdl-36357670

ABSTRACT

Alcohol use disorder (AUD) is a chronic and fatal disease. The main impediment of the AUD therapy is a high probability of relapse to alcohol abuse even after prolonged abstinence. The molecular mechanisms of cue-induced relapse are not well established, despite the fact that they may offer new targets for the treatment of AUD. Using a comprehensive animal model of AUD, virally-mediated and amygdala-targeted genetic manipulations by CRISPR/Cas9 technology and ex vivo electrophysiology, we identify a mechanism that selectively controls cue-induced alcohol relapse and AUD symptom severity. This mechanism is based on activity-regulated cytoskeleton-associated protein (Arc)/ARG3.1-dependent plasticity of the amygdala synapses. In humans, we identified single nucleotide polymorphisms in the ARC gene and their methylation predicting not only amygdala size, but also frequency of alcohol use, even at the onset of regular consumption. Targeting Arc during alcohol cue exposure may thus be a selective new mechanism for relapse prevention.


Subject(s)
Alcoholism , Central Amygdaloid Nucleus , Animals , Humans , Alcoholism/genetics , Chronic Disease , Cues , Ethanol , Recurrence , Nerve Tissue Proteins/metabolism , Cytoskeletal Proteins/metabolism
5.
Mol Psychiatry ; 27(3): 1694-1703, 2022 03.
Article in English | MEDLINE | ID: mdl-34997193

ABSTRACT

The amygdala, a critical brain region responsible for emotional behavior, is crucially involved in the regulation of the effects of stress on emotional behavior. In the mammalian forebrain, gastrin-releasing peptide (GRP), a 27-amino-acid mammalian neuropeptide, which is a homolog of the 14-amino-acid amidated amphibian peptide bombesin, is highly expressed in the amygdala. The levels of GRP are markedly increased in the amygdala after acute stress; therefore, it is known as a stress-activated modulator. To determine the role of GRP in emotional behavior under stress, we conducted some behavioral and biochemical experiments with GRP-knockout (KO) mice. GRP-KO mice exhibited a longer freezing response than wild-type (WT) littermates in both contextual and auditory fear (also known as threat) conditioning tests only when they were subjected to acute restraint stress 20 min before the conditioning. To identify the critical neural circuits associated with the regulation of emotional memory by GRP, we conducted Arc/Arg3.1-reporter mapping in the amygdala with an Arc-Venus reporter transgenic mouse line. In the amygdalostriatal transition area (AST) and the lateral side of the basal nuclei, fear conditioning after restraint stress increased neuronal activity significantly in WT mice, and GRP KO was found to negate this potentiation only in the AST. These results indicate that the GRP-activated neurons in the AST are likely to suppress excessive fear expression through the regulation of downstream circuits related to fear learning following acute stress.


Subject(s)
Bombesin , Fear , Amygdala/metabolism , Animals , Bombesin/metabolism , Bombesin/pharmacology , Conditioning, Classical/physiology , Fear/physiology , Gastrin-Releasing Peptide/metabolism , Gastrin-Releasing Peptide/pharmacology , Mammals/metabolism , Mice , Mice, Knockout
6.
Nat Methods ; 16(11): 1119-1122, 2019 11.
Article in English | MEDLINE | ID: mdl-31659327

ABSTRACT

Two-photon microscopy is a mainstay technique for imaging in scattering media and normally provides frame-acquisition rates of ~10-30 Hz. To track high-speed phenomena, we created a two-photon microscope with 400 illumination beams that collectively sample 95,000-211,000 µm2 areas at rates up to 1 kHz. Using this microscope, we visualized microcirculatory flow, fast venous constrictions and neuronal Ca2+ spiking with millisecond-scale timing resolution in the brains of awake mice.


Subject(s)
Brain/blood supply , Microscopy, Fluorescence, Multiphoton/methods , Animals , Calcium/metabolism , Male , Mice , Mice, Inbred C57BL , Microcirculation , Wakefulness
7.
Mol Psychiatry ; 26(2): 710-720, 2021 02.
Article in English | MEDLINE | ID: mdl-30262887

ABSTRACT

A discrepancy in oxytocin's behavioral effects between acute and repeated administrations indicates distinct underlying neurobiological mechanisms. The current study employed a combination of human clinical trial and animal study to compare neurochemical changes induced by acute and repeated oxytocin administrations. Human study analyzed medial prefrontal metabolite levels by using 1H-magnetic resonance spectroscopy, a secondary outcome in our randomized, double-blind, placebo-controlled crossover trial of 6 weeks intranasal administrations of oxytocin (48 IU/day) and placebo within-subject design in 17 psychotropic-free high-functioning men with autism spectrum disorder. Medial prefrontal transcript expression levels were analyzed in adult male C57BL/6J mice after intraperitoneal injection of oxytocin or saline either once (200 ng/100 µL/mouse, n = 12) or for 14 consecutive days (200 ng/100 µL/mouse/day, n = 16). As the results, repeated administration of oxytocin significantly decreased the medial prefrontal N-acetylaspartate (NAA; p = 0.043) and glutamate-glutamine levels (Glx; p = 0.001), unlike the acute oxytocin. The decreases were inversely and specifically associated (r = 0.680, p = 0.004 for NAA; r = 0.491, p = 0.053 for Glx) with oxytocin-induced improvements of medial prefrontal functional MRI activity during a social judgment task not with changes during placebo administrations. In wild-type mice, we found that repeated oxytocin administration reduced medial frontal transcript expression of N-methyl-D-aspartate receptor type 2B (p = 0.018), unlike the acute oxytocin, which instead changed the transcript expression associated with oxytocin (p = 0.0004) and neural activity (p = 0.0002). The present findings suggest that the unique sensitivity of the glutamatergic system to repeated oxytocin administration may explain the differential behavioral effects of oxytocin between acute and repeated administration.


Subject(s)
Autism Spectrum Disorder , Oxytocin , Administration, Intranasal , Animals , Autism Spectrum Disorder/drug therapy , Double-Blind Method , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Oxytocin/therapeutic use
8.
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
9.
J Biol Chem ; 295(23): 7923-7940, 2020 06 05.
Article in English | MEDLINE | ID: mdl-32341124

ABSTRACT

RNA localization in subcellular compartments is essential for spatial and temporal regulation of protein expression in neurons. Several techniques have been developed to visualize mRNAs inside cells, but the study of the behavior of endogenous and nonengineered mRNAs in living neurons has just started. In this study, we combined reduction-triggered fluorescent (RETF) probes and fluorescence correlation spectroscopy (FCS) to investigate the diffusion properties of activity-regulated cytoskeleton-associated protein (Arc) and inositol 1,4,5-trisphosphate receptor type 1 (Ip3r1) mRNAs. This approach enabled us to discriminate between RNA-bound and unbound fluorescent probes and to quantify mRNA diffusion parameters and concentrations in living rat primary hippocampal neurons. Specifically, we detected the induction of Arc mRNA production after neuronal activation in real time. Results from computer simulations with mRNA diffusion coefficients obtained in these analyses supported the idea that free diffusion is incapable of transporting mRNA of sizes close to those of Arc or Ip3r1 to distal dendrites. In conclusion, the combined RETF-FCS approach reported here enables analyses of the dynamics of endogenous, unmodified mRNAs in living neurons, affording a glimpse into the intracellular dynamics of RNA in live cells.


Subject(s)
Fluorescent Dyes/chemistry , Neurons/chemistry , RNA, Messenger/metabolism , Animals , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Fluorescent Dyes/chemical synthesis , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Optical Imaging , RNA, Messenger/genetics , Rats , Rats, Wistar , Spectrometry, Fluorescence
10.
Semin Cell Dev Biol ; 77: 43-50, 2018 05.
Article in English | MEDLINE | ID: mdl-28939038

ABSTRACT

Long-lasting forms of synaptic plasticity such as long-term potentiation (LTP) and long-term depression (LTD) are fundamental cellular mechanisms underlying learning and memory. The synaptic tagging and capture (STC) hypothesis has provided a theoretical framework on how products of activity-dependent genes may interact with potentiated synapses to facilitate and maintain such long-lasting synaptic plasticity. Although Arc/arg3.1 was initially assumed to participate in STC processes during LTP, accumulating evidence indicated that Arc/arg3.1 might rather contribute in weakening of synaptic weights than in their strengthening. In particular, analyses of Arc/Arg3.1 protein dynamics and function in the dendrites after plasticity-inducing stimuli have revealed a new type of inactivity-dependent redistribution of synaptic weights, termed "inverse synaptic tagging". The original synaptic tagging and inverse synaptic tagging likely co-exist and are mutually non-exclusive mechanisms, which together may help orchestrate the redistribution of synaptic weights and promote the enhancement and maintenance of their contrast between potentiated and non-potentiated synapses during the late phase of long-term synaptic plasticity. In this review, we describe the inverse synaptic tagging mechanism that controls synaptic dynamics of Arc/Arg3.1, an immediate early gene product which is captured and preferentially targeted to non-potentiated synapses, and discuss its impact on neuronal circuit refinement and cognitive function.


Subject(s)
Cognition/physiology , Cytoskeletal Proteins/metabolism , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Synapses/metabolism , Humans , Learning/physiology , Memory/physiology , Proteins/metabolism , Receptors, Glutamate/metabolism
11.
Proc Natl Acad Sci U S A ; 114(25): 6599-6604, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28584117

ABSTRACT

Aberrant histaminergic function has been proposed as a cause of tic disorders. A rare mutation in the enzyme that produces histamine (HA), histidine decarboxylase (HDC), has been identified in patients with Tourette syndrome (TS). Hdc knockout mice exhibit repetitive behavioral pathology and neurochemical characteristics of TS, establishing them as a plausible model of tic pathophysiology. Where, when, and how HA deficiency produces these effects has remained unclear: whether the contribution of HA deficiency to pathogenesis is acute or developmental, and where in the brain the relevant consequences of HA deficiency occur. Here, we address these key pathophysiological questions, using anatomically and cellularly targeted manipulations in mice. We report that specific ablation or chemogenetic silencing of histaminergic neurons in the tuberomammillary nucleus (TMN) of the hypothalamus leads to markedly elevated grooming, a form of repetitive behavioral pathology, and to elevated markers of neuronal activity in both dorsal striatum and medial prefrontal cortex. Infusion of HA directly into the striatum reverses this behavioral pathology, confirming that acute HA deficiency mediates the effect. Bidirectional chemogenetic regulation reveals that dorsal striatum neurons activated after TMN silencing are both sufficient to produce repetitive behavioral pathology and necessary for the full expression of the effect. Chemogenetic activation of TMN-regulated medial prefrontal cortex neurons, in contrast, increases locomotion and not grooming. These data confirm the centrality of striatal regulation by neurotransmitter HA in the adult in the production of pathological grooming.


Subject(s)
Basal Ganglia/metabolism , Grooming/physiology , Histamine/metabolism , Animals , Corpus Striatum/metabolism , Histidine Decarboxylase/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Prefrontal Cortex/metabolism , Spinal Cord Dorsal Horn/metabolism , Tourette Syndrome/metabolism
12.
Proc Natl Acad Sci U S A ; 114(34): 9182-9187, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28790183

ABSTRACT

The molecular basis for the decline in experience-dependent neural plasticity over age remains poorly understood. In visual cortex, the robust plasticity induced in juvenile mice by brief monocular deprivation during the critical period is abrogated by genetic deletion of Arc, an activity-dependent regulator of excitatory synaptic modification. Here, we report that augmenting Arc expression in adult mice prolongs juvenile-like plasticity in visual cortex, as assessed by recordings of ocular dominance (OD) plasticity in vivo. A distinguishing characteristic of juvenile OD plasticity is the weakening of deprived-eye responses, believed to be accounted for by the mechanisms of homosynaptic long-term depression (LTD). Accordingly, we also found increased LTD in visual cortex of adult mice with augmented Arc expression and impaired LTD in visual cortex of juvenile mice that lack Arc or have been treated in vivo with a protein synthesis inhibitor. Further, we found that although activity-dependent expression of Arc mRNA does not change with age, expression of Arc protein is maximal during the critical period and declines in adulthood. Finally, we show that acute augmentation of Arc expression in wild-type adult mouse visual cortex is sufficient to restore juvenile-like plasticity. Together, our findings suggest a unifying molecular explanation for the age- and activity-dependent modulation of synaptic sensitivity to deprivation.


Subject(s)
Cytoskeletal Proteins/physiology , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Visual Cortex/physiology , Age Factors , Animals , Cytoskeletal Proteins/genetics , Dominance, Ocular/genetics , Dominance, Ocular/physiology , Gene Expression Regulation, Developmental , Long-Term Synaptic Depression/genetics , Long-Term Synaptic Depression/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neuronal Plasticity/genetics , Visual Cortex/growth & development , Visual Cortex/metabolism
13.
J Neurosci ; 38(24): 5551-5566, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29773754

ABSTRACT

Despite many association studies linking gene polymorphisms and mutations of L-type voltage-gated Ca2+ channels (VGCCs) in neurodevelopmental disorders such as autism and schizophrenia, the roles of specific L-type VGCC during brain development remain unclear. Calcium signaling has been shown to be essential for neurodevelopmental processes such as sculpting of neurites, functional wiring, and fine tuning of growing networks. To investigate this relationship, we performed submembraneous calcium imaging using a membrane-tethered genetically encoded calcium indicator (GECI) Lck-G-CaMP7. We successfully recorded spontaneous regenerative calcium transients (SRCaTs) in developing mouse excitatory cortical neurons prepared from both sexes before synapse formation. SRCaTs originated locally in immature neurites independently of somatic calcium rises and were significantly more elevated in the axons than in dendrites. SRCaTs were not blocked by tetrodoxin, a Na+ channel blocker, but were strongly inhibited by hyperpolarization, suggesting a voltage-dependent source. Pharmacological and genetic manipulations revealed the critical importance of the Cav1.2 (CACNA1C) pore-forming subunit of L-type VGCCs, which were indeed expressed in immature mouse brains. Consistently, knocking out Cav1.2 resulted in significant alterations of neurite outgrowth. Furthermore, expression of a gain-of-function Cav1.2 mutant found in Timothy syndrome, an autosomal dominant multisystem disorder exhibiting syndromic autism, resulted in impaired radial migration of layer 2/3 excitatory neurons, whereas postnatal abrogation of Cav1.2 enhancement could rescue cortical malformation. Together, these lines of evidence suggest a critical role for spontaneous opening of L-type VGCCs in neural development and corticogenesis and indicate that L-type VGCCs might constitute a perinatal therapeutic target for neuropsychiatric calciochannelopathies.SIGNIFICANCE STATEMENT Despite many association studies linking gene polymorphisms and mutations of L-type voltage-gated Ca2+ channels (VGCCs) in neurodevelopmental disorders such as autism and schizophrenia, the roles of specific L-type VGCCs during brain development remain unclear. We here combined the latest Ca2+ indicator technology, quantitative pharmacology, and in utero electroporation and found a hitherto unsuspected role for L-type VGCCs in determining the Ca2+ signaling landscape of mouse immature neurons. We found that malfunctional L-type VGCCs in immature neurons before birth might cause errors in neuritic growth and cortical migration. Interestingly, the retarded corticogenesis phenotype was rescued by postnatal correction of L-type VGCC signal aberration. These findings suggest that L-type VGCCs might constitute a perinatal therapeutic target for neurodevelopment-associated psychiatric disorders.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Signaling/physiology , Cerebral Cortex/growth & development , Neurogenesis/physiology , Neuronal Outgrowth/physiology , Animals , Cell Movement/physiology , Cerebral Cortex/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Neural Stem Cells/metabolism
14.
J Neurochem ; 148(2): 204-218, 2019 01.
Article in English | MEDLINE | ID: mdl-30244496

ABSTRACT

The expression of immediate early genes (IEGs) is thought to be an essential molecular basis of neuronal plasticity for higher brain function. Many IEGs contain serum response element in their transcriptional regulatory regions and their expression is controlled by serum response factor (SRF). SRF is known to play a role in concert with transcriptional cofactors. However, little is known about how SRF cofactors regulate IEG expression during the process of neuronal plasticity. We hypothesized that one of the SRF-regulated neuronal IEGs, activity-regulated cytoskeleton-associated protein (Arc; also termed Arg3.1), is regulated by an SRF coactivator, megakaryoblastic leukemia (MKL). To test this hypothesis, we initially investigated which binding site of the transcription factor or SRF cofactor contributes to brain-derived neurotrophic factor (BDNF)-induced Arc gene transcription in cultured cortical neurons using transfection and reporter assays. We found that BDNF caused robust induction of Arc gene transcription through a cAMP response element, binding site of myocyte enhancer factor 2, and binding site of SRF in an Arc enhancer, the synaptic activity-responsive element (SARE). Regardless of the requirement for the SRF-binding site, the binding site of a ternary complex factor, another SRF cofactor, did not affect BDNF-mediated Arc gene transcription. In contrast, chromatin immunoprecipitation revealed occupation of MKL at the SARE. Furthermore, knockdown of MKL2, but not MKL1, significantly decreased BDNF-mediated activation of the SARE. Taken together, these findings suggest a novel mechanism by which MKL2 controls the Arc SARE in response to BDNF stimulation.


Subject(s)
Cytoskeletal Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neurons/physiology , Transcription Factors/metabolism , Transcriptional Activation/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Cytoskeletal Proteins/genetics , Female , Nerve Tissue Proteins/genetics , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Serum Response Factor/genetics , Serum Response Factor/metabolism , Transcriptional Activation/drug effects
15.
Nat Methods ; 13(4): 325-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26878381

ABSTRACT

Real-time activity measurements from multiple specific cell populations and projections are likely to be important for understanding the brain as a dynamical system. Here we developed frame-projected independent-fiber photometry (FIP), which we used to record fluorescence activity signals from many brain regions simultaneously in freely behaving mice. We explored the versatility of the FIP microscope by quantifying real-time activity relationships among many brain regions during social behavior, simultaneously recording activity along multiple axonal pathways during sensory experience, performing simultaneous two-color activity recording, and applying optical perturbation tuned to elicit dynamics that match naturally occurring patterns observed during behavior.


Subject(s)
Brain Mapping/methods , Brain/physiology , Calcium Signaling , Neural Pathways , Photometry/methods , Social Behavior , Animals , Brain/cytology , Mice
16.
Mol Ther ; 26(10): 2397-2406, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30064895

ABSTRACT

In patients born blind with retinal dystrophies, understanding the critical periods of cortical plasticity is important for successful visual restoration. In this study, we sought to model childhood blindness and investigate the plasticity of visual pathways. To this end, we generated double-mutant (Pde6ccpfl1/cpfl1Gnat1IRD2/IRD2) mice with absent rod and cone photoreceptor function, and we evaluated their response for restoring rod (GNAT1) function through gene therapy. Despite the limited effectiveness of gene therapy in restoring visual acuity in patients with retinal dystrophy, visual acuity was, unexpectedly, successfully restored in the mice at the level of the primary visual cortex in this study. This success in visual restoration, defined by changes in the quantified optokinetic response and pattern visually evoked potential, was achieved regardless of the age at treatment (up to 16 months). In the contralateral visual cortex, cortical plasticity, tagged with light-triggered transcription of Arc, was also restored after the treatment in blind mice carrying an Arc promoter-driven reporter gene, dVenus. Our results demonstrate the remarkable plasticity of visual circuits for one of the two photoreceptor mechanisms in older as well as younger mice with congenital blindness due to retinal dystrophies.


Subject(s)
Blindness/therapy , GTP-Binding Protein alpha Subunits/genetics , Genetic Therapy , Retinal Dystrophies/therapy , Transducin/genetics , Visual Acuity/genetics , Animals , Blindness/genetics , Blindness/pathology , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Electroretinography , GTP-Binding Protein alpha Subunits/administration & dosage , Humans , Mice , Mutation , Retina/drug effects , Retina/pathology , Retinal Cone Photoreceptor Cells/drug effects , Retinal Cone Photoreceptor Cells/pathology , Retinal Dystrophies/genetics , Retinal Dystrophies/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Transducin/administration & dosage , Visual Acuity/drug effects , Visual Cortex/drug effects , Visual Cortex/pathology
17.
J Neurochem ; 146(2): 145-159, 2018 07.
Article in English | MEDLINE | ID: mdl-29675826

ABSTRACT

Drebrin is a major F-actin binding protein in dendritic spines that is critically involved in the regulation of dendritic spine morphogenesis, pathology, and plasticity. In this study, we aimed to identify a novel drebrin-binding protein involved in spine morphogenesis and synaptic plasticity. We confirmed the beta subunit of Ca2+ /calmodulin-dependent protein kinase II (CaMKIIß) as a drebrin-binding protein using a yeast two-hybrid system, and investigated the drebrin-CaMKIIß relationship in dendritic spines using rat hippocampal neurons. Drebrin knockdown resulted in diffuse localization of CaMKIIß in dendrites during the resting state, suggesting that drebrin is involved in the accumulation of CaMKIIß in dendritic spines. Fluorescence recovery after photobleaching analysis showed that drebrin knockdown increased the stable fraction of CaMKIIß, indicating the presence of drebrin-independent, more stable CaMKIIß. NMDA receptor activation also increased the stable fraction in parallel with drebrin exodus from dendritic spines. These findings suggest that CaMKIIß can be classified into distinct pools: CaMKIIß associated with drebrin, CaMKIIß associated with post-synaptic density (PSD), and CaMKIIß free from PSD and drebrin. CaMKIIß appears to be anchored to a protein complex composed of drebrin-binding F-actin during the resting state. NMDA receptor activation releases CaMKIIß from drebrin resulting in CaMKIIß association with PSD.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dendrites/ultrastructure , Dendritic Spines/metabolism , Neurons/cytology , Neuropeptides/metabolism , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Chlorocebus aethiops , Embryo, Mammalian , Excitatory Amino Acid Agonists/pharmacology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glutamic Acid/pharmacology , Glycine/pharmacology , Hippocampus/cytology , Neuropeptides/genetics , Photobleaching , Pregnancy , Protein Binding/drug effects , Protein Binding/genetics , Pseudopodia/drug effects , Pseudopodia/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Transgenic , Rats, Wistar
18.
Biochem Biophys Res Commun ; 505(4): 1216-1222, 2018 11 10.
Article in English | MEDLINE | ID: mdl-30322614

ABSTRACT

Astrocytes are known to contact with a great number of synapses and may integrate sensory inputs. In the ferret primary visual cortex, astrocytes respond to a visual stimulus with a delay of several seconds with respect to the surrounding neurons. However, in the mouse visual cortex, it remains unclear whether astrocytes respond to visual stimulations. In this study, using dual-color simultaneous in vivo two-photon calcium imaging of neurons and astrocytes in the awake mouse visual cortex, we examined the visual response of astrocytes and their precise response timing relative to the surrounding neurons. Neurons reliably responded to visual stimulations, whereas astrocytes often showed neuromodulator-mediated global activities, which largely masked small visual responses. Administration of the selective α1-adrenergic receptor antagonist prazosin substantially reduced such global astrocytic activities without affecting the neuronal visual responses. In the presence of prazosin, astrocytes showed weak but consistent visual responses mostly at their somata. Cross-correlation analysis estimated that the astrocytic visual responses were delayed by approximately 5 seconds relative to the surrounding neuronal responses. In conclusion, our research demonstrated that astrocytes in the primary visual cortex of awake mice responded to visual stimuli with a delay of several seconds relative to the surrounding neurons, which may indicate the existence of a common mechanism of neuron-astrocyte communication across species.


Subject(s)
Astrocytes/metabolism , Visual Cortex/metabolism , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Animals , Astrocytes/drug effects , Calcium/metabolism , Male , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Photic Stimulation , Prazosin/pharmacology , Visual Cortex/cytology
19.
Nat Methods ; 12(1): 64-70, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25419959

ABSTRACT

Fluorescent Ca(2+) reporters are widely used as readouts of neuronal activities. Here we designed R-CaMP2, a high-affinity red genetically encoded calcium indicator (GECI) with a Hill coefficient near 1. Use of the calmodulin-binding sequence of CaMKK-α and CaMKK-ß in lieu of an M13 sequence resulted in threefold faster rise and decay times of Ca(2+) transients than R-CaMP1.07. These features allowed resolving single action potentials (APs) and recording fast AP trains up to 20-40 Hz in cortical slices. Somatic and synaptic activities of a cortical neuronal ensemble in vivo were imaged with similar efficacy as with previously reported sensitive green GECIs. Combining green and red GECIs, we successfully achieved dual-color monitoring of neuronal activities of distinct cell types, both in the mouse cortex and in freely moving Caenorhabditis elegans. Dual imaging using R-CaMP2 and green GECIs provides a powerful means to interrogate orthogonal and hierarchical neuronal ensembles in vivo.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Indicators and Reagents/chemical synthesis , Action Potentials/physiology , Animals , Caenorhabditis elegans/radiation effects , Calcium/metabolism , Calcium Signaling/physiology , Calmodulin-Binding Proteins , Cells, Cultured , Cerebral Cortex/cytology , Fluorescent Dyes/metabolism , HEK293 Cells , Hippocampus/cytology , Humans , Light , Mice , Neurons/physiology , Patch-Clamp Techniques , Peptide Fragments/chemistry , Peptide Fragments/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL