Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Nat Commun ; 15(1): 4531, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38866749

ABSTRACT

Individuals with autism spectrum disorder (ASD) have a higher prevalence of social memory impairment. A series of our previous studies revealed that hippocampal ventral CA1 (vCA1) neurons possess social memory engram and that the neurophysiological representation of social memory in the vCA1 neurons is disrupted in ASD-associated Shank3 knockout mice. However, whether the dysfunction of Shank3 in vCA1 causes the social memory impairment observed in ASD remains unclear. In this study, we found that vCA1-specific Shank3 conditional knockout (cKO) by the adeno-associated virus (AAV)- or specialized extracellular vesicle (EV)- mediated in vivo gene editing was sufficient to recapitulate the social memory impairment in male mice. Furthermore, the utilization of EV-mediated Shank3-cKO allowed us to quantitatively examine the role of Shank3 in social memory. Our results suggested that there is a certain threshold for the proportion of Shank3-cKO neurons required for social memory disruption. Thus, our study provides insight into the population coding of social memory in vCA1, as well as the pathological mechanisms underlying social memory impairment in ASD.


Subject(s)
Autism Spectrum Disorder , CA1 Region, Hippocampal , Gene Editing , Memory , Mice, Knockout , Nerve Tissue Proteins , Social Behavior , Animals , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , CA1 Region, Hippocampal/metabolism , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Mice , Memory/physiology , Neurons/metabolism , Dependovirus/genetics , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Memory Disorders/genetics , Memory Disorders/metabolism , Memory Disorders/physiopathology , Mice, Inbred C57BL
2.
Cancer Sci ; 115(6): 1924-1935, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38549229

ABSTRACT

In childhood acute lymphoblastic leukemia (ALL), TP53 gene mutation is associated with chemoresistance in a certain population of relapsed cases. To directly verify the association of TP53 gene mutation with chemoresistance of relapsed childhood ALL cases and improve their prognosis, the development of appropriate human leukemia models having TP53 mutation in the intrinsic gene is required. Here, we sought to introduce R248Q hotspot mutation into the intrinsic TP53 gene in an ALL cell line, 697, by applying a prime editing (PE) system, which is a versatile genome editing technology. The PE2 system uses an artificial fusion of nickase Cas9 and reverse-transcriptase to directly place new genetic information into a target site through a reverse transcriptase template in the prime editing guide RNA (pegRNA). Moreover, in the advanced PE3b system, single guide RNA (sgRNA) matching the edited sequence is also introduced to improve editing efficiency. The initially obtained MDM2 inhibitor-resistant PE3b-transfected subline revealed disrupted p53 transactivation activity, reduced p53 target gene expression, and acquired resistance to chemotherapeutic agents and irradiation. Although the majority of the subline acquired the designed R248Q and adjacent silent mutations, the insertion of the palindromic sequence in the scaffold hairpin structure of pegRNA and the overlap of the original genomic DNA sequence were frequently observed. Targeted next-generation sequencing reconfirmed frequent edit errors in both PE2 and PE3b-transfected 697 cells, and it revealed frequent successful edits in HEK293T cells. These observations suggest a requirement for further modification of the PE2 and PE3b systems for accurate editing in leukemic cells.


Subject(s)
Gene Editing , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/genetics , Gene Editing/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , CRISPR-Cas Systems/genetics , RNA, Guide, CRISPR-Cas Systems/genetics , Proto-Oncogene Proteins c-mdm2/genetics
3.
Nat Neurosci ; 27(3): 561-572, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38243089

ABSTRACT

Episodic memories are encoded by experience-activated neuronal ensembles that remain necessary and sufficient for recall. However, the temporal evolution of memory engrams after initial encoding is unclear. In this study, we employed computational and experimental approaches to examine how the neural composition and selectivity of engrams change with memory consolidation. Our spiking neural network model yielded testable predictions: memories transition from unselective to selective as neurons drop out of and drop into engrams; inhibitory activity during recall is essential for memory selectivity; and inhibitory synaptic plasticity during memory consolidation is critical for engrams to become selective. Using activity-dependent labeling, longitudinal calcium imaging and a combination of optogenetic and chemogenetic manipulations in mouse dentate gyrus, we conducted contextual fear conditioning experiments that supported our model's predictions. Our results reveal that memory engrams are dynamic and that changes in engram composition mediated by inhibitory plasticity are crucial for the emergence of memory selectivity.


Subject(s)
Memory Consolidation , Memory, Episodic , Mice , Animals , Memory Consolidation/physiology , Mental Recall/physiology , Neurons/physiology , Fear/physiology
4.
J Neurochem ; 165(2): 211-229, 2023 04.
Article in English | MEDLINE | ID: mdl-36807153

ABSTRACT

Astrotactin2 (ASTN2) regulates neuronal migration and synaptic strength through the trafficking and degradation of surface proteins. Deletion of ASTN2 in copy number variants has been identified in patients with schizophrenia, bipolar disorder, and autism spectrum disorder in copy number variant (CNV) analysis. Disruption of ASTN2 is a risk factor for these neurodevelopmental disorders, including schizophrenia, bipolar disorder, autism spectrum disorder, and attention deficit hyperactivity disorder. However, the importance of ASTN2 in physiological functions remains poorly understood. To elucidate the physiological functions of ASTN2, we investigated whether deficiency of ASTN2 affects cognitive and/or emotional behaviors and neurotransmissions using ASTN2-deficient mice. Astn2 knockout (KO) mice produced by CRISPR/Cas9 technique showed no obvious differences in physical characteristics and circadian rhythm. Astn2 KO mice showed increased exploratory activity in a novel environment, social behavior and impulsivity, or decreased despair-, anxiety-like behaviors and exploratory preference for the novel object. Some behavioral abnormalities, such as increased exploratory activity and impulsivity, or decreased exploratory preference were specifically attenuated by risperidone, but not by haloperidol. While, the both drugs did not affect any emotion-related behavioral abnormalities in Astn2 KO mice. Dopamine contents were decreased in the striatum, and serotonin or dopamine turnover were increased in the striatum, nucleus accumbens, and amygdala of Astn2 KO mice. In morphological analyses, thinning of neural cell layers in the hippocampus, reduction of neural cell bodies in the prefrontal cortex, and decrease in spine density and PSD95 protein in both tissues were observed in Astn2 KO mice. The present findings suggest that ASTN2 deficiency develops some emotional or cognitive impairments related to monoaminergic dysfunctions and abnormal neuronal morphogenesis with shrinkage of neuronal soma. ASTN2 protein may contribute to the pathogenic mechanism and symptom onset of mental disorders.


Subject(s)
Autism Spectrum Disorder , Dopamine , Animals , Mice , Cognition , Dopamine/metabolism , Emotions , Glycoproteins/metabolism , Mice, Knockout , Morphogenesis
5.
Nat Neurosci ; 25(11): 1458-1469, 2022 11.
Article in English | MEDLINE | ID: mdl-36319770

ABSTRACT

Synaptic pruning is a fundamental process of neuronal circuit refinement in learning and memory. Accumulating evidence suggests that glia participates in sculpting the neuronal circuits through synapse engulfment. However, whether glial involvement in synaptic pruning has a role in memory formation remains elusive. Using newly developed phagocytosis reporter mice and three-dimensional ultrastructural characterization, we found that synaptic engulfment by cerebellar Bergmann glia (BG) frequently occurred upon cerebellum-dependent motor learning in mice. We observed increases in pre- and postsynaptic nibbling by BG along with a reduction in spine volume after learning. Pharmacological blockade of engulfment with Annexin V inhibited both the spine volume reduction and overnight improvement of motor adaptation. These results indicate that BG contribute to the refinement of the mature cerebellar cortical circuit through synaptic engulfment during motor learning.


Subject(s)
Neuroglia , Synapses , Mice , Animals , Neuroglia/physiology , Synapses/physiology , Neurons/physiology , Cerebellum/physiology , Neuronal Plasticity
6.
eNeuro ; 9(5)2022.
Article in English | MEDLINE | ID: mdl-35977823

ABSTRACT

Fragile X syndrome (FXS) is a leading monogenic cause of intellectual disability and autism spectrum disorders, spurring decades of intense research and a multitude of mouse models. So far, these models do not recapitulate the genetic underpinning of classical FXS-CGG repeat-induced methylation of the Fmr1 locus-and their findings have failed to translate into the clinic. We sought to answer whether this disparity was because of low repeat length and generated a novel mouse line with 341 repeats, Fmr1hs341 , which is the largest allele in mice reported to date. This repeat length is significantly longer than the 200 repeats generally required for methylation of the repeat tract and promoter region in FXS patients, which leads to silencing of the FMR1 gene. Bisulfite sequencing fails to detect the robust methylation expected of FXS in Fmr1hs341 mice. Quantitative real-time PCR and Western blotting results also do not resemble FXS and instead produce a biochemical profile consistent with the fragile X-associated premutation disorders. These findings suggest that repeat length is unlikely to be the core determinant preventing methylation in mice, and other organisms phylogenetically closer to humans may be required to effectively model FXS.


Subject(s)
Fragile X Mental Retardation Protein , Fragile X Syndrome , Animals , DNA Methylation , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Humans , Mice , Trinucleotide Repeat Expansion/genetics
7.
Nature ; 607(7918): 321-329, 2022 07.
Article in English | MEDLINE | ID: mdl-35676479

ABSTRACT

Although bradykinesia, tremor and rigidity are the hallmark motor defects in patients with Parkinson's disease (PD), patients also experience motor learning impairments and non-motor symptoms such as depression1. The neural circuit basis for these different symptoms of PD are not well understood. Although current treatments are effective for locomotion deficits in PD2,3, therapeutic strategies targeting motor learning deficits and non-motor symptoms are lacking4-6. Here we found that distinct parafascicular (PF) thalamic subpopulations project to caudate putamen (CPu), subthalamic nucleus (STN) and nucleus accumbens (NAc). Whereas PF→CPu and PF→STN circuits are critical for locomotion and motor learning, respectively, inhibition of the PF→NAc circuit induced a depression-like state. Whereas chemogenetically manipulating CPu-projecting PF neurons led to a long-term restoration of locomotion, optogenetic long-term potentiation (LTP) at PF→STN synapses restored motor learning behaviour in an acute mouse model of PD. Furthermore, activation of NAc-projecting PF neurons rescued depression-like phenotypes. Further, we identified nicotinic acetylcholine receptors capable of modulating PF circuits to rescue different PD phenotypes. Thus, targeting PF thalamic circuits may be an effective strategy for treating motor and non-motor deficits in PD.


Subject(s)
Affect , Motor Skills , Neural Pathways , Parkinson Disease , Thalamus , Animals , Disease Models, Animal , Learning , Locomotion , Long-Term Potentiation , Mice , Neurons/physiology , Nucleus Accumbens , Optogenetics , Parkinson Disease/physiopathology , Parkinson Disease/psychology , Parkinson Disease/therapy , Putamen , Receptors, Nicotinic , Subthalamic Nucleus , Synapses , Thalamus/cytology , Thalamus/pathology
8.
Proc Natl Acad Sci U S A ; 119(20): e2118712119, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35537049

ABSTRACT

Alterations in the structure and functional connectivity of anterior thalamic nuclei (ATN) have been linked to reduced cognition during aging. However, ATN circuits that contribute to higher cognitive functions remain understudied. We found that the anteroventral (AV) subdivision of ATN is necessary specifically during the maintenance phase of a spatial working memory task. This function engages the AV→parasubiculum (PaS)→entorhinal cortex (EC) circuit. Aged mice showed a deficit in spatial working memory, which was associated with a decrease in the excitability of AV neurons. Activation of AV neurons or the AV→PaS circuit in aged mice was sufficient to rescue their working memory performance. Furthermore, rescued aged mice showed improved behavior-induced neuronal activity in prefrontal cortex (PFC), a critical site for working memory processes. Although the direct activation of PFC neurons in aged mice also rescued their working memory performance, we found that these animals exhibited increased levels of anxiety, which was not the case for AV→PaS circuit manipulations in aged mice. These results suggest that targeting AV thalamus in aging may not only be beneficial for cognitive functions but that this approach may have fewer unintended effects compared to direct PFC manipulations.


Subject(s)
Anterior Thalamic Nuclei , Animals , Anterior Thalamic Nuclei/physiology , Cognition , Memory Disorders , Memory, Short-Term/physiology , Mice , Neural Pathways/physiology , Neurons
9.
Nat Commun ; 13(1): 1799, 2022 04 04.
Article in English | MEDLINE | ID: mdl-35379803

ABSTRACT

Neuronal ensembles that hold specific memory (memory engrams) have been identified in the hippocampus, amygdala, or cortex. However, it has been hypothesized that engrams of a specific memory are distributed among multiple brain regions that are functionally connected, referred to as a unified engram complex. Here, we report a partial map of the engram complex for contextual fear conditioning memory by characterizing encoding activated neuronal ensembles in 247 regions using tissue phenotyping in mice. The mapping was aided by an engram index, which identified 117 cFos+ brain regions holding engrams with high probability, and brain-wide reactivation of these neuronal ensembles by recall. Optogenetic manipulation experiments revealed engram ensembles, many of which were functionally connected to hippocampal or amygdala engrams. Simultaneous chemogenetic reactivation of multiple engram ensembles conferred a greater level of memory recall than reactivation of a single engram ensemble, reflecting the natural memory recall process. Overall, our study supports the unified engram complex hypothesis for memory storage.


Subject(s)
Brain Mapping , Memory , Animals , Brain , Fear/physiology , Hippocampus/physiology , Memory/physiology , Mice
10.
Gut ; 71(3): 487-496, 2022 03.
Article in English | MEDLINE | ID: mdl-33963042

ABSTRACT

OBJECTIVE: Although immunoglobulin A (IgA) is abundantly expressed in the gut and known to be an important component of mucosal barriers against luminal pathogens, its precise function remains unclear. Therefore, we tried to elucidate the effect of IgA on gut homeostasis maintenance and its mechanism. DESIGN: We generated various IgA mutant mouse lines using the CRISPR/Cas9 genome editing system. Then, we evaluated the effect on the small intestinal homeostasis, pathology, intestinal microbiota, cytokine production, and immune cell activation using intravital imaging. RESULTS: We obtained two lines, with one that contained a <50 base pair deletion in the cytoplasmic region of the IgA allele (IgA tail-mutant; IgAtm/tm) and the other that lacked the most constant region of the IgH α chain, which resulted in the deficiency of IgA production (IgA-/-). IgA-/- exhibited spontaneous inflammation in the ileum but not the other parts of the gastrointestinal tract. Associated with this, there were significantly increased lamina propria CD4+ T cells, elevated productions of IFN-γ and IL-17, increased ileal segmented filamentous bacteria and skewed intestinal microflora composition. Intravital imaging using Ca2+ biosensor showed that IgA-/- had elevated Ca2+ signalling in Peyer's patch B cells. On the other hand, IgAtm/tm seemed to be normal, suggesting that the IgA cytoplasmic tail is dispensable for the prevention of the intestinal disorder. CONCLUSION: IgA plays an important role in the mucosal homeostasis associated with the regulation of intestinal microbiota and protection against mucosal inflammation especially in the ileum.


Subject(s)
Ileitis/etiology , Ileum/pathology , Immunoglobulin A/physiology , Animals , B-Lymphocytes/physiology , Cytokines/metabolism , Disease Models, Animal , Female , Gastrointestinal Microbiome , Homeostasis , Ileitis/metabolism , Ileitis/pathology , Ileum/metabolism , Ileum/ultrastructure , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Intravital Microscopy , Male , Mice , Mice, Mutant Strains , T-Lymphocytes/physiology
11.
Neurochem Int ; 150: 105177, 2021 11.
Article in English | MEDLINE | ID: mdl-34481039

ABSTRACT

The importance of glutamate transporters in learning, memory, and emotion remains poorly understood; hence, in the present study, we investigated whether deficiency of pharmacological GLAST in neurodevelopmental processes affects cognitive and/or emotional behaviors in mice. The mice were injected with a glutamate transporter inhibitor, dl-threo-ß-benzyloxyaspartate (dl-TBOA), during the early postnatal period. At 8 weeks of age, they showed impairments in cognitive or emotional behaviors; dysfunction of glutamatergic neurotransmission (increased expressions of GLAST, GLT-1, or GFAP protein, and decreased ability of glutamate release) in the cortex or hippocampus; morphological changes (decreased cell size in the cortex and thickness of the pyramidal neuronal layer of the CA1 area in the hippocampus). Such behavioral and morphological changes were not observed in adult mice injected with dl-TBOA. These results suggest that GLAST plays an important role in the regulation of cognitive and emotional behaviors. Early postnatal glutamatergic facilitation by GLAST dysfunction leads to cognitive and emotional abnormalities due to neurodevelopmental abnormalities such as morphological changes.


Subject(s)
Aspartic Acid/toxicity , Excitatory Amino Acid Transporter 1/antagonists & inhibitors , Excitatory Amino Acid Transporter 1/metabolism , Mental Disorders/chemically induced , Mental Disorders/metabolism , Neurons/metabolism , Animals , Animals, Newborn , Aspartic Acid/administration & dosage , Female , Injections, Intraventricular , Male , Maze Learning/drug effects , Maze Learning/physiology , Mental Disorders/pathology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/pathology , Pregnancy
12.
Cell Rep ; 36(5): 109492, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34348144

ABSTRACT

Early differential diagnosis between malignant and benign tumors and their underlying intrinsic differences are the most critical issues for life-threatening cancers. To study whether human acral melanomas, deadly cancers that occur on non-hair-bearing skin, have distinct origins that underlie their invasive capability, we develop fate-tracing technologies of melanocyte stem cells in sweat glands (glandular McSCs) and in melanoma models in mice and compare the cellular dynamics with human melanoma. Herein, we report that glandular McSCs self-renew to expand their migratory progeny in response to genotoxic stress and trauma to generate invasive melanomas in mice that mimic human acral melanomas. The analysis of melanocytic lesions in human volar skin reveals that genetically unstable McSCs expand in sweat glands and in the surrounding epidermis in melanomas but not in nevi. The detection of such cell spreading dynamics provides an innovative method for an early differential diagnosis of acral melanomas from nevi.


Subject(s)
Cell Movement , Melanoma/pathology , Nevus/pathology , Stem Cells/pathology , Animals , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Cyclin D1/metabolism , Disease Models, Animal , Epidermis/pathology , Epidermis/radiation effects , Gene Amplification , Genomic Instability/radiation effects , Melanocytes/pathology , Melanocytes/radiation effects , Melanoma/diagnosis , Mice, Inbred C57BL , Risk Factors , Skin/pathology , Skin/radiation effects , Skin Pigmentation/radiation effects , Sweat Glands/radiation effects , Ultraviolet Rays
13.
Neuron ; 109(16): 2590-2603.e13, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34197733

ABSTRACT

Neuropsychiatric disorders are often accompanied by cognitive impairments/intellectual disability (ID). It is not clear whether there are converging mechanisms underlying these debilitating impairments. We found that many autism and schizophrenia risk genes are expressed in the anterodorsal subdivision (AD) of anterior thalamic nuclei, which has reciprocal connectivity with learning and memory structures. CRISPR-Cas9 knockdown of multiple risk genes selectively in AD thalamus led to memory deficits. While the AD is necessary for contextual memory encoding, the neighboring anteroventral subdivision (AV) regulates memory specificity. These distinct functions of AD and AV are mediated through their projections to retrosplenial cortex, using differential mechanisms. Furthermore, knockdown of autism and schizophrenia risk genes PTCHD1, YWHAG, or HERC1 from AD led to neuronal hyperexcitability, and normalization of hyperexcitability rescued memory deficits in these models. This study identifies converging cellular to circuit mechanisms underlying cognitive deficits in a subset of neuropsychiatric disease models.


Subject(s)
Anterior Thalamic Nuclei/physiopathology , Cognitive Dysfunction/physiopathology , Neural Pathways/physiopathology , Thalamic Nuclei/physiopathology , Animals , Anterior Thalamic Nuclei/physiology , Cerebral Cortex/physiopathology , Cognition/physiology , Mice , Neural Pathways/physiology , Thalamic Nuclei/physiology
14.
Cell ; 184(12): 3267-3280.e18, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34043941

ABSTRACT

Searching for factors to improve knockin efficiency for therapeutic applications, biotechnology, and generation of non-human primate models of disease, we found that the strand exchange protein RAD51 can significantly increase Cas9-mediated homozygous knockin in mouse embryos through an interhomolog repair (IHR) mechanism. IHR is a hallmark of meiosis but only occurs at low frequencies in somatic cells, and its occurrence in zygotes is controversial. Using multiple approaches, we provide evidence for an endogenous IHR mechanism in the early embryo that can be enhanced by RAD51. This process can be harnessed to generate homozygotes from wild-type zygotes using exogenous donors and to convert heterozygous alleles into homozygous alleles without exogenous templates. Furthermore, we identify additional IHR-promoting factors and describe features of IHR events. Together, our findings show conclusive evidence for IHR in mouse embryos and describe an efficient method for enhanced gene conversion.


Subject(s)
DNA Repair/genetics , Gene Conversion , Rad51 Recombinase/metabolism , Alleles , Animals , Base Sequence , CRISPR-Associated Protein 9/metabolism , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Chromosomes, Mammalian/genetics , DNA Breaks, Double-Stranded , Embryo, Mammalian , Female , Genetic Loci , Homologous Recombination/genetics , Homozygote , Humans , INDEL Mutation/genetics , Mice, Inbred C57BL , Mosaicism , Nuclear Proteins/metabolism , Polymorphism, Single Nucleotide/genetics , Ribonucleoproteins/metabolism , Zygote/metabolism
15.
J Neurosci ; 41(10): 2106-2118, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33478985

ABSTRACT

Seizures invite seizures. At the initial stage of epilepsy, seizures intensify with each episode; however, the mechanisms underlying this exacerbation remain to be solved. Astrocytes have a strong control over neuronal excitability and the mode of information processing. This control is accomplished by adjusting the levels of various ions in the extracellular space. The network of astrocytes connected via gap junctions allows a wider or more confined distribution of these ions depending on the open probability of the gap junctions. K+ clearance relies on the K+ uptake by astrocytes and the subsequent diffusion of K+ through the astrocyte network. When astrocytes become uncoupled, K+ clearance becomes hindered. Accumulation of extracellular K+ leads to hyperexcitability of neurons. Here, using acute hippocampal slices from mice, we uncovered that brief periods of epileptiform activity result in gap junction uncoupling. In slices that experienced short-term epileptiform activity, extracellular K+ transients in response to glutamate became prolonged. Na+ imaging with a fluorescent indicator indicated that intercellular diffusion of small cations in the astrocytic syncytium via gap junctions became rapidly restricted after epileptiform activity. Using a transgenic mouse with astrocyte-specific expression of a pH sensor (Lck-E2GFP), we confirmed that astrocytes react to epileptiform activity with intracellular alkalization. Application of Na+/HCO3- cotransporter blocker led to the suppression of intracellular alkalization of astrocytes and to the prevention of astrocyte uncoupling and hyperactivity intensification both in vitro and in vivo Therefore, the inhibition of astrocyte alkalization could become a promising therapeutic strategy for countering epilepsy development.SIGNIFICANCE STATEMENT We aimed to understand the mechanisms underlying the plastic change of forebrain circuits associated with the intensification of epilepsy. Here, we demonstrate that first-time exposure to only brief periods of epileptiform activity results in acute disturbance of the intercellular astrocyte network formed by gap junctions in hippocampal tissue slices from mice. Moreover, rapid clearance of K+ from the extracellular space was impaired. Epileptiform activity activated inward Na+/HCO3- cotransport in astrocytes by cell depolarization, resulting in their alkalization. Our data suggest that alkaline pH shifts in astrocytes lead to gap junction uncoupling, hampering K+ clearance, and thereby to exacerbation of epilepsy. Pharmacological intervention could become a promising new strategy to dampen neuronal hyperexcitability and epileptogenesis.


Subject(s)
Astrocytes/metabolism , Epilepsy/metabolism , Epilepsy/physiopathology , Gap Junctions/metabolism , Animals , Hippocampus , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Neurons/metabolism , Potassium/metabolism
16.
Biosci Microbiota Food Health ; 39(4): 209-218, 2020.
Article in English | MEDLINE | ID: mdl-33117619

ABSTRACT

Mechanosensory systems have been implicated in the maintenance of gut homeostasis, but details on the related mechanisms are scarce. Recently, we generated a conditional Ca2+ biosensor yellow cameleon 3.60 (YC3.60)-expressing transgenic mouse model and established a five-dimensional (5D; x, y, z, time, and Ca2+) intravital imaging system for investigating lymphoid tissues and enteric epithelial cell responses. To validate this gut-sensing system, we visualized responses of enteric nervous system (ENS) cells in Nestin-Cre/YC3.60flox mice with specific YC3.60 expression. The ENS, including the myenteric (Auerbach's) and submucous (Meissner's) plexuses, could be visualized without staining in this mouse line, indicating that the probe produced sufficient fluorescent intensity. Furthermore, the myenteric plexus exhibited Ca2+ signaling during peristalsis without stimulation. Nerve endings on the surface of enteric epithelia also exhibited Ca2+ signaling without stimulation. Mechanical stress induced transient salient Ca2+ flux in the myenteric plexus and in enteric epithelial cells in the Nestin-Cre/YC3.60 and the CAG-Cre/YC3.60 lines, respectively. Furthermore, the potential TRPM7 inhibitors were shown to attenuate mechanical stress-mediated Ca2+ signaling. These data indicate that the present intravital imaging system can be used to visualize mechanosensory Ca2+ signaling in ENS cells and enteric epithelial cells.

17.
Neuroimage ; 223: 117318, 2020 12.
Article in English | MEDLINE | ID: mdl-32882386

ABSTRACT

Diffusion functional magnetic resonance imaging (DfMRI) has been proposed as an alternative functional imaging method to detect brain activity without confounding hemodynamic effects. Here, taking advantage of this DfMRI feature, we investigated abnormalities of dynamic brain function in a neuropsychiatric disease mouse model (glial glutamate transporter-knockdown mice with obsessive-compulsive disorder [OCD]-related behavior). Our DfMRI approaches consisted of three analyses: resting state brain activity, functional connectivity, and propagation of neural information. We detected hyperactivation and biased connectivity across the cortico-striatal-thalamic circuitry, which is consistent with known blood oxygen-level dependent (BOLD)-fMRI patterns in OCD patients. In addition, we performed ignition-driven mean integration (IDMI) analysis, which combined activity and connectivity analyses, to evaluate neural propagation initiated from brain activation. This analysis revealed an unbalanced distribution of neural propagation initiated from intrinsic local activation to the global network, while these were not detected by the conventional method with BOLD-fMRI. This abnormal function detected by DfMRI was associated with OCD-related behavior. Together, our comprehensive DfMRI approaches can successfully provide information on dynamic brain function in normal and diseased brains.


Subject(s)
Brain/pathology , Brain/physiopathology , Diffusion Magnetic Resonance Imaging , Obsessive-Compulsive Disorder/pathology , Obsessive-Compulsive Disorder/physiopathology , Animals , Brain/diagnostic imaging , Brain Mapping/methods , Disease Models, Animal , Excitatory Amino Acid Transporter 2/genetics , Gene Knockdown Techniques , Mice , Neural Pathways/diagnostic imaging , Neural Pathways/pathology , Neural Pathways/physiopathology , Obsessive-Compulsive Disorder/diagnostic imaging
18.
J Neurosci ; 40(38): 7241-7254, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32847967

ABSTRACT

Maladaptation to stress is a critical risk factor in stress-related disorders, such as major depression and post-traumatic stress disorder (PTSD). Dopamine signaling in the nucleus accumbens (NAc) has been shown to modulate behavior by reinforcing learning and evading aversive stimuli, which are important for the survival of animals under environmental challenges such as stress. However, the mechanisms through which dopaminergic transmission responds to stressful events and subsequently regulates its downstream neuronal activity during stress remain unknown. To investigate how dopamine signaling modulates stress-coping behavior, we measured the subsecond fluctuation of extracellular dopamine concentration and pH using fast scanning cyclic voltammetry (FSCV) in the NAc, a postsynaptic target of midbrain dopaminergic neurons, in male mice engaged in a tail suspension test (TST). The results revealed a transient decrease in dopamine concentration and an increase in pH levels when the animals changed behaviors, from being immobile to struggling. Interestingly, optogenetic inhibition of dopamine release in NAc, potentiated the struggling behavior in animals under the TST. We then addressed the causal relationship of such a dopaminergic transmission with behavioral alterations by knocking out both the dopamine receptors, i.e., D1 and D2, in the NAc using viral vector-mediated genome editing. Behavioral analyses revealed that male D1 knock-out mice showed significantly more struggling bouts and longer struggling durations during the TST, while male D2 knock-out mice did not. Our results therefore indicate that D1 dopaminergic signaling in the NAc plays a pivotal role in the modulation of stress-coping behaviors in animals under tail suspension stress.SIGNIFICANCE STATEMENT The tail suspension test (TST) has been widely used as a despair-based behavioral assessment to screen the antidepressant so long. Despite its prevalence in the animal studies, the neural substrate underlying the changes of behavior during the test remains unclear. This study provides an evidence for a role of dopaminergic transmission in the modulation of stress-coping behavior during the TST, a despair test widely used to screen the antidepressants in rodents. Taking into consideration the fact that the dopamine metabolism is upregulated by almost all antidepressants, a part of which acts directly on the dopaminergic transmission, current results would uncover the molecular mechanism through which the dopaminergic signaling mediates antidepressant effect with facilitation of the recovery from the despair-like behavior in the TST.


Subject(s)
Adaptation, Psychological , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Nucleus Accumbens/metabolism , Stress, Psychological/metabolism , Animals , Cell Line, Tumor , Dopaminergic Neurons/physiology , Male , Mice , Mice, Inbred C57BL , Nucleus Accumbens/cytology , Nucleus Accumbens/physiopathology , Receptors, Dopamine/genetics , Receptors, Dopamine/metabolism , Stress, Psychological/physiopathology , Synaptic Transmission
19.
Biochem Biophys Res Commun ; 529(4): 943-949, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32819603

ABSTRACT

Glaucoma is one of the leading causes of blindness characterized by progressive loss of retinal ganglion cells (RGCs) and their axons. We reported that glutamate/aspartate transporter (GLAST) knockout mice showed progressive RGC loss and optic nerve degeneration that are similar to glaucoma. To explore the possibility that rare variants in the EAAT1 gene (the human homolog of GLAST) cause susceptibility to glaucoma, we performed targeted sequencing of EAAT1 in 440 patients with glaucoma and 450 control subjects. We identified 8 rare variants in 20 out of 440 patients, including 4 synonymous and 4 missense variants located at protein coding regions. One of these rare variants (rs117295512) showed significant association with the risk of glaucoma (OR = 10.44, P = 0.005). Furthermore, the allele frequency for loss-of-function EAAT1 variants, pAla169Gly and pAla329Thr, was 5.5 folds higher in the glaucoma (1.1%) compared with the control cohort (0.2%). These findings suggest that these rare variants may contribute to the pathogenesis of glaucoma and that loss-of-function variants in EAAT1 are present in a small number of patients with glaucoma.


Subject(s)
Excitatory Amino Acid Transporter 1/genetics , Glaucoma, Open-Angle/genetics , Low Tension Glaucoma/genetics , Mutation, Missense , Silent Mutation , Alleles , Amino Acid Sequence , Animals , Case-Control Studies , Cell Line , Excitatory Amino Acid Transporter 1/deficiency , Gene Expression , Gene Frequency , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/pathology , Humans , Intraocular Pressure , Low Tension Glaucoma/metabolism , Low Tension Glaucoma/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Optic Nerve/metabolism , Optic Nerve/pathology , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , Risk Factors , Sequence Alignment , Sequence Homology, Amino Acid
20.
Nature ; 583(7818): 819-824, 2020 07.
Article in English | MEDLINE | ID: mdl-32699411

ABSTRACT

The thalamic reticular nucleus (TRN), the major source of thalamic inhibition, regulates thalamocortical interactions that are critical for sensory processing, attention and cognition1-5. TRN dysfunction has been linked to sensory abnormality, attention deficit and sleep disturbance across multiple neurodevelopmental disorders6-9. However, little is known about the organizational principles that underlie its divergent functions. Here we performed an integrative study linking single-cell molecular and electrophysiological features of the mouse TRN to connectivity and systems-level function. We found that cellular heterogeneity in the TRN is characterized by a transcriptomic gradient of two negatively correlated gene-expression profiles, each containing hundreds of genes. Neurons in the extremes of this transcriptomic gradient express mutually exclusive markers, exhibit core or shell-like anatomical structure and have distinct electrophysiological properties. The two TRN subpopulations make differential connections with the functionally distinct first-order and higher-order thalamic nuclei to form molecularly defined TRN-thalamus subnetworks. Selective perturbation of the two subnetworks in vivo revealed their differential role in regulating sleep. In sum, our study provides a comprehensive atlas of TRN neurons at single-cell resolution and links molecularly defined subnetworks to the functional organization of thalamocortical circuits.


Subject(s)
Gene Regulatory Networks , Thalamic Nuclei/cytology , Thalamic Nuclei/metabolism , Animals , Cluster Analysis , Female , Gene Expression Profiling , In Situ Hybridization, Fluorescence , Metalloendopeptidases/metabolism , Mice , Neural Pathways , Neurons/metabolism , Osteopontin/metabolism , Patch-Clamp Techniques , RNA-Seq , Single-Cell Analysis , Sleep/genetics , Sleep/physiology , Thalamic Nuclei/physiology , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...