Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 27(1): 116-128, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38012399

ABSTRACT

Whole-brain genome editing to correct single-base mutations and reduce or reverse behavioral changes in animal models of autism spectrum disorder (ASD) has not yet been achieved. We developed an apolipoprotein B messenger RNA-editing enzyme, catalytic polypeptide-embedded cytosine base editor (AeCBE) system for converting C·G to T·A base pairs. We demonstrate its effectiveness by targeting AeCBE to an ASD-associated mutation of the MEF2C gene (c.104T>C, p.L35P) in vivo in mice. We first constructed Mef2cL35P heterozygous mice. Male heterozygous mice exhibited hyperactivity, repetitive behavior and social abnormalities. We then programmed AeCBE to edit the mutated C·G base pairs of Mef2c in the mouse brain through the intravenous injection of blood-brain barrier-crossing adeno-associated virus. This treatment successfully restored Mef2c protein levels in several brain regions and reversed the behavioral abnormalities in Mef2c-mutant mice. Our work presents an in vivo base-editing paradigm that could potentially correct single-base genetic mutations in the brain.


Subject(s)
Autism Spectrum Disorder , Gene Editing , Animals , Mice , Male , Autism Spectrum Disorder/genetics , Brain , Mutation/genetics , MEF2 Transcription Factors/genetics
3.
Elife ; 102021 06 29.
Article in English | MEDLINE | ID: mdl-34184986

ABSTRACT

The congenital intellectual disability (ID)-causing gene mutations remain largely unclear, although many genetic variations might relate to ID. We screened gene mutations in Chinese Han children suffering from severe ID and found a single-nucleotide polymorphism (SNP) in the 5'-untranslated region (5'-UTR) of fibroblast growth factor 13 (FGF13) mRNA (NM_001139500.1:c.-32c>G) shared by three male children. In both HEK293 cells and patient-derived induced pluripotent stem cells, this SNP reduced the translation of FGF13, which stabilizes microtubules in developing neurons. Mice carrying the homologous point mutation in 5'-UTR of Fgf13 showed delayed neuronal migration during cortical development, and weakened learning and memory. Furthermore, this SNP reduced the interaction between FGF13 5'-UTR and polypyrimidine-tract-binding protein 2 (PTBP2), which was required for FGF13 translation in cortical neurons. Thus, this 5'-UTR SNP of FGF13 interferes with the translational process of FGF13 and causes deficits in brain development and cognitive functions.


Subject(s)
5' Untranslated Regions/genetics , Fibroblast Growth Factors/genetics , Intellectual Disability/genetics , Point Mutation , Polymorphism, Single Nucleotide , Adolescent , Animals , Child , Child, Preschool , Fibroblast Growth Factors/metabolism , HEK293 Cells , Humans , Learning , Male , Memory , Mice , Mice, Inbred C57BL
4.
Sci Bull (Beijing) ; 66(9): 937-946, 2021 May 15.
Article in English | MEDLINE | ID: mdl-36654241

ABSTRACT

Although CRISPR/Cas9-mediated gene editing is widely applied to mimic human disorders, whether acute manipulation of disease-causing genes in the brain leads to behavioral abnormalities in non-human primates remains to be determined. Here we induced genetic mutations in MECP2, a critical gene linked to Rett syndrome (RTT) and autism spectrum disorders (ASD), in the hippocampus (DG and CA1-4) of adolescent rhesus monkeys (Macaca mulatta) in vivo via adeno-associated virus (AAV)-delivered Staphylococcus aureus Cas9 with small guide RNAs (sgRNAs) targeting MECP2. In comparison to monkeys injected with AAV-SaCas9 alone (n = 4), numerous autistic-like behavioral abnormalities were identified in the AAV-SaCas9-sgMECP2-injected monkeys (n = 7), including social interaction deficits, abnormal sleep patterns, insensitivity to aversive stimuli, abnormal hand motions, and defective social reward behaviors. Furthermore, some aspects of ASD and RTT, such as stereotypic behaviors, did not appear in the MECP2 gene-edited monkeys, suggesting that different brain areas likely contribute to distinct ASD symptoms. This study showed that acute manipulation of disease-causing genes via in vivo gene editing directly led to behavioral changes in adolescent primates, paving the way for the rapid generation of genetically engineered non-human primate models for neurobiological studies and therapeutic development.

5.
Neurosci Bull ; 36(6): 570-584, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32144612

ABSTRACT

Methyl-CpG binding protein 2 (MeCP2) is a basic nuclear protein involved in the regulation of gene expression and microRNA processing. Duplication of MECP2-containing genomic segments causes MECP2 duplication syndrome, a severe neurodevelopmental disorder characterized by intellectual disability, motor dysfunction, heightened anxiety, epilepsy, autistic phenotypes, and early death. Reversal of the abnormal phenotypes in adult mice with MECP2 duplication (MECP2-TG) by normalizing the MeCP2 levels across the whole brain has been demonstrated. However, whether different brain areas or neural circuits contribute to different aspects of the behavioral deficits is still unknown. Here, we found that MECP2-TG mice showed a significant social recognition deficit, and were prone to display aversive-like behaviors, including heightened anxiety-like behaviors and a fear generalization phenotype. In addition, reduced locomotor activity was observed in MECP2-TG mice. However, appetitive behaviors and learning and memory were comparable in MECP2-TG and wild-type mice. Functional magnetic resonance imaging illustrated that the differences between MECP2-TG and wild-type mice were mainly concentrated in brain areas regulating emotion and social behaviors. We used the CRISPR-Cas9 method to restore normal MeCP2 levels in the medial prefrontal cortex (mPFC) and bed nuclei of the stria terminalis (BST) of adult MECP2-TG mice, and found that normalization of MeCP2 levels in the mPFC but not in the BST reversed the social recognition deficit. These data indicate that the mPFC is responsible for the social recognition deficit in the transgenic mice, and provide new insight into potential therapies for MECP2 duplication syndrome.


Subject(s)
Methyl-CpG-Binding Protein 2 , Prefrontal Cortex , Recognition, Psychology , Social Behavior , Animals , Anxiety , China , Disease Models, Animal , Fear , Gene Duplication , Male , Methyl-CpG-Binding Protein 2/metabolism , Mice , Mice, Transgenic , Prefrontal Cortex/metabolism
6.
Cell Death Differ ; 26(10): 1863-1879, 2019 10.
Article in English | MEDLINE | ID: mdl-30560934

ABSTRACT

Duplication of MECP2 (Methyl-CpG-binding protein 2) causes severe mental illness called MECP2 duplication syndrome (MDS), yet the underlying mechanism remains elusive. Here we show, in Tg(MECP2) transgenic mouse brain or cultured neural progenitor cells (NPCs), that elevated MeCP2 expression promotes NPC differentiation into neurons. Ectopic expression of MeCP2 inhibits ADAM10 and thus the NOTCH pathway during NPC differentiation. In human cells, this downregulation on ADAM10 was mediated by miRNA-197, which is upregulated by MeCP2. Surprisingly, miR-197 binds to the ADAM10 3'-UTR via its 3' side, not the canonical seed sequence on the 5' side. In mouse cells, a noncoding RNA Gm28836 is used to replace the function of miR-197 between MeCP2 and ADAM10. Similar to MeCP2, overexpressing miR-197 also promotes NPCs differentiation into neurons. Interestingly, three rare missense mutations (H371R, E394K, and G428S) in MECP2, which we identified in a Han Chinese autism spectrum disorders (ASD) cohort showed loss-of-function effects in NPC differentiation assay. These mutations cannot upregulate miR-197. Overexpressing miR-197 together with these MeCP2 mutations could rescue the downregulation on ADAM10. Not only the inhibitor of miR-197 could reverse the effect of overexpressed MeCP2 on NPCs differentiation, but also overexpression of miR-197 could reverse the NPCs differentiation defects caused by MECP2 mutations. Our results revealed that a regulatory axis involving MeCP2, miR-197, ADAM10, and NOTCH signaling is critical for NPC differentiation, which is affected by both MeCP2 duplication and mutation.


Subject(s)
ADAM10 Protein/biosynthesis , Amyloid Precursor Protein Secretases/biosynthesis , Cell Differentiation , Gene Expression Regulation, Enzymologic , Membrane Proteins/biosynthesis , Methyl-CpG-Binding Protein 2/metabolism , MicroRNAs/metabolism , Neural Stem Cells/metabolism , ADAM10 Protein/genetics , Amyloid Precursor Protein Secretases/genetics , Animals , Asian People , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Cell Line , China , Humans , Membrane Proteins/genetics , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Transgenic , MicroRNAs/genetics , Mutation, Missense , Neural Stem Cells/pathology
7.
Cancer Med ; 7(12): 6147-6157, 2018 12.
Article in English | MEDLINE | ID: mdl-30402980

ABSTRACT

Circular RNAs (circRNAs) have been demonstrated to be involved in various biological processes. Nevertheless, the function of circRNAs in medulloblastoma (MB) is still unknown. The present study aimed to investigate the expression profiles of circRNAs and related mechanisms for regulating the proliferation and growth of tumor cells in MB. The expression profiles of circRNAs were screened from four normal cerebellum and four MB samples using a HiSeq Sequencer. Bioinformatic analysis was employed to predict the interaction between circRNAs and mRNAs in MB. Subsequently, the expression levels of eight differential circRNAs [circ-SKA3 (hsa_circ_0029696), circ-DTL (hsa_circ_0000179), circ-CRTAM, circ-MAP3K5 (hsa_circ_0006856), circ-RIMS1-1 (hsa_circ_0132250), circ-RIMS1-2 (hsa_circ_0076967), circ-FLT3-1 (hsa_circ_0100165), and circ-FLT3-2 (hsa_circ_0100168)] were validated using quantitative reverse transcription-polymerase chain reaction. Moreover, circ-SKA3 and circ-DTL were silenced using small interfering RNAs and their host genes were overexpressed to investigate their role in the pathogenesis of MB. A total of 33 circRNAs were found to be differentially expressed in MB tissues (fold change ≥ 2.0, FDR <0.05), of which three were upregulated and 30 were downregulated; six circRNAs were experimentally validated successfully. Upregulated circ-SKA3 and circ-DTL promoted the proliferation migration and invasion in vitro by regulating the expression of host genes. This novel study exploited the profiling of circRNAs in MB and demonstrated that circ-SKA3 and circ-DTL were crucial in the tumorigenesis and development of MB and might be considered as novel and potential biomarkers for the diagnosis and new targets for the intervention of MB.


Subject(s)
Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , RNA , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cerebellar Neoplasms/pathology , Cerebellum/metabolism , Down-Regulation , Humans , Medulloblastoma/pathology , RNA, Circular , RNA, Messenger , Up-Regulation
8.
Mol Med Rep ; 18(6): 5016-5022, 2018 12.
Article in English | MEDLINE | ID: mdl-30280194

ABSTRACT

Retinitis pigmentosa (RP) is a common form of inherited retinal degeneration that causes progressive loss of vision or adult blindness, characterized by the impairment of rod and cone photoreceptors. At present, mutations in >60 pathogenic genes have been confirmed to cause RP. The predominant modes of inheritance are autosomal dominant, autosomal recessive and X­linked. In addition, other modes of inheritance, including digenic or mitochondrial inheritance, have been reported. In previous decades, with the development of sequencing techniques, significant advances in identifying novel RP pathogenic genes and screening mutations have been made. In the present study, whole­exome sequencing was performed on samples from two Chinese pedigrees diagnosed with RP. A compound heterozygous mutation in the gene usherin 2A (USH2A; c.6,485+5G>A/c.11,156G>A) and a heterozygous X­linked mutation in the gene retinitis pigmentosa 2 (RP2) ARL3 GTPase­activating protein (RP2; c.358C>T) were identified by Sanger sequencing and co­segregation analysis, of which the pathogenic mutation (c.6,485+5G>A) in USH2A has not been previously reported among Chinese patients. The findings of the present study may expand on current knowledge of RP among the Chinese population, providing essential assistance in the molecular diagnosis and screening of RP, and promoting further investigation of the pathogenesis of RP.


Subject(s)
Extracellular Matrix Proteins/genetics , Genome, Human , Mutation , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Whole Genome Sequencing , Adult , Alleles , China , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Pedigree , Phenotype , Tomography, Optical Coherence
9.
Nat Commun ; 7: 12229, 2016 08 11.
Article in English | MEDLINE | ID: mdl-27510304

ABSTRACT

The coexistence of electrical and chemical synapses among interneurons is essential for interneuron function in the neocortex. However, it remains largely unclear whether electrical coupling between interneurons influences chemical synapse formation and microcircuit assembly during development. Here, we show that electrical and GABAergic chemical connections robustly develop between interneurons in neocortical layer 1 over a similar time course. Electrical coupling promotes action potential generation and synchronous firing between layer 1 interneurons. Furthermore, electrically coupled interneurons exhibit strong GABA-A receptor-mediated synchronous synaptic activity. Disruption of electrical coupling leads to a loss of bidirectional, but not unidirectional, GABAergic connections. Moreover, a reduction in electrical coupling induces an increase in excitatory synaptic inputs to layer 1 interneurons. Together, these findings strongly suggest that electrical coupling between neocortical interneurons plays a critical role in regulating chemical synapse development and precise formation of circuits.


Subject(s)
Interneurons/physiology , Neocortex/embryology , Neocortex/physiology , Action Potentials/physiology , Animals , Connexins/physiology , Gap Junctions/physiology , Mice , Neural Inhibition/physiology , RNA Interference , Receptors, GABA-A/metabolism , Synapses/physiology , Synaptic Potentials , gamma-Aminobutyric Acid/physiology , Gap Junction delta-2 Protein
11.
Sci Rep ; 6: 20392, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26843422

ABSTRACT

MeCP2 encodes a methyl-CpG-binding protein that plays a critical role in repressing gene expression, mutations of which lead to Rett syndrome and autism. PTEN is a critical tumor suppressor gene that is frequently mutated in human cancers and autism spectrum disorders. Various studies have shown that both MeCP2 and PTEN proteins play important roles in brain development. Here we find that MeCP2 and PTEN reciprocally regulate expression of each other via microRNAs. Knockdown of MeCP2 leads to upregulation of microRNA-137, which in turn represses expression of PTEN, thus PTEN would be down-regulated when MeCP2 is knockdown. Furthermore, we find that deletion of PTEN leads to phosphorylation of Serine 133 of CREB, then increases the expression of microRNA-132. miR-132 inhibits the expression of MeCP2 by targeting on the 3'UTR of MeCP2 mRNA. Our work shows that two critical disorders-related gene MeCP2 and PTEN reciprocally regulate expression of each other by distinct mechanisms, suggesting that rare mutations in various disorders may lead to dysregulation of other critical genes and yield unexpected consequences.


Subject(s)
Methyl-CpG-Binding Protein 2/metabolism , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , 3' Untranslated Regions , Animals , Autistic Disorder/genetics , Autistic Disorder/pathology , Blotting, Western , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Down-Regulation , Humans , Methyl-CpG-Binding Protein 2/antagonists & inhibitors , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Neurons/cytology , Neurons/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , Phosphorylation , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Sequence Analysis, RNA , Up-Regulation
12.
Neuropsychopharmacology ; 41(8): 2133-46, 2016 07.
Article in English | MEDLINE | ID: mdl-26797244

ABSTRACT

The basal forebrain (BF) cholinergic neurons have long been thought to be involved in behavioral wakefulness and cortical activation. However, owing to the heterogeneity of BF neurons and poor selectivity of traditional methods, the precise role of BF cholinergic neurons in regulating the sleep-wake cycle remains unclear. We investigated the effects of cell-selective manipulation of BF cholinergic neurons on the sleep-wake behavior and electroencephalogram (EEG) power spectrum using the pharmacogenetic technique, the 'designer receptors exclusively activated by designer drugs (DREADD)' approach, and ChAT-IRES-Cre mice. Our results showed that activation of BF cholinergic neurons expressing hM3Dq receptors significantly and lastingly decreased the EEG delta power spectrum, produced low-delta non-rapid eye movement sleep, and slightly increased wakefulness in both light and dark phases, whereas inhibition of BF cholinergic neurons expressing hM4Di receptors significantly increased EEG delta power spectrum and slightly decreased wakefulness. Next, the projections of BF cholinergic neurons were traced by humanized Renilla green fluorescent protein (hrGFP). Abundant and highly dense hrGFP-positive fibers were observed in the secondary motor cortex and cingulate cortex, and sparse hrGFP-positive fibers were observed in the ventrolateral preoptic nucleus, a known sleep-related structure. Finally, we found that activation of BF cholinergic neurons significantly increased c-Fos expression in the secondary motor cortex and cingulate cortex, but decreased c-Fos expression in the ventrolateral preoptic nucleus. Taken together, these findings reveal that the primary function of BF cholinergic neurons is to inhibit EEG delta activity through the activation of cerebral cortex, rather than to induce behavioral wakefulness.


Subject(s)
Basal Forebrain/physiology , Cholinergic Neurons/physiology , Delta Rhythm , Sleep , Wakefulness , Animals , Basal Forebrain/cytology , Cerebral Cortex/cytology , Cholinergic Neurons/cytology , Electroencephalography , Male , Mice , Mice, Transgenic , Neural Pathways/cytology , Neural Pathways/physiology , Sleep Stages
14.
Mol Brain ; 8: 39, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26104391

ABSTRACT

BACKGROUND: Young neurons in the developing brain establish a polarized morphology for proper migration. The PIWI family of piRNA processing proteins are considered to be restrictively expressed in germline tissues and several types of cancer cells. They play important roles in spermatogenesis, stem cell maintenance, piRNA biogenesis, and transposon silencing. Interestingly a recent study showed that de novo mutations of PIWI family members are strongly associated with autism. RESULTS: Here, we report that PIWI-like 1 (PIWIL1), a PIWI family member known to be essential for the transition of round spermatid into elongated spermatid, plays a role in the polarization and radial migration of newborn neurons in the developing cerebral cortex. Knocking down PIWIL1 in newborn cortical neurons by in utero electroporation of specific siRNAs resulted in retardation of the transition of neurons from the multipolar stage to the bipolar stage followed by a defect in their radial migration to the proper destination. Domain analysis showed that both the RNA binding PAZ domain and the RNA processing PIWI domain in PIWIL1 were indispensable for its function in neuronal migration. Furthermore, we found that PIWIL1 unexpectedly regulates the expression of microtubule-associated proteins in cortical neurons. CONCLUSIONS: PIWIL1 regulates neuronal polarization and radial migration partly via modulating the expression of microtubule-associated proteins (MAPs). Our finding of PIWIL1's function in neuronal development implies conserved functions of molecules participating in morphogenesis of brain and germline tissue and provides a mechanism as to how mutations of PIWI may be associated with autism.


Subject(s)
Argonaute Proteins/metabolism , Cell Movement , Cell Polarity , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Animals , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , DNA Methylation/genetics , Gene Knockdown Techniques , Humans , Mice, Inbred C57BL , Mitosis , Neurons/metabolism , Protein Structure, Tertiary , RNA Stability , Rats, Sprague-Dawley
15.
PLoS One ; 10(4): e0123585, 2015.
Article in English | MEDLINE | ID: mdl-25894199

ABSTRACT

Neural stem cell (NSC) proliferation and differentiation are required to replace neurons damaged or lost after hypoxic-ischemic events and recover brain function. Periostin (POSTN), a novel matricellular protein, plays pivotal roles in the survival, migration, and regeneration of various cell types, but its function in NSCs of neonatal rodent brain is still unknown. The purpose of this study was to investigate the role of POSTN in NSCs following hypoxia-ischemia (HI). We found that POSTN mRNA levels significantly increased in differentiating NSCs. The proliferation and differentiation of NSCs in the hippocampus is compromised in POSTN knockout mice. Moreover, NSC proliferation and differentiation into neurons and astrocytes significantly increased in cultured NSCs treated with recombinant POSTN. Consistently, injection of POSTN into neonatal hypoxic-ischemic rat brains stimulated NSC proliferation and differentiation in the subventricular and subgranular zones after 7 and 14 days of brain injury. Lastly, POSTN treatment significantly improved the spatial learning deficits of rats subjected to HI. These results suggest that POSTN significantly enhances NSC proliferation and differentiation after HI, and provides new insights into therapeutic strategies for the treatment of hypoxic-ischemic encephalopathy.


Subject(s)
Brain Ischemia/pathology , Cell Adhesion Molecules/metabolism , Cell Differentiation , Neural Stem Cells/metabolism , Animals , Animals, Newborn , Brain Ischemia/complications , Brain Ischemia/genetics , Cell Adhesion Molecules/genetics , Cell Hypoxia , Cell Proliferation , Cognition Disorders/complications , Female , Male , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley
16.
PLoS One ; 10(3): e0115997, 2015.
Article in English | MEDLINE | ID: mdl-25790286

ABSTRACT

Periventricular leukomalacia (PVL) is a common ischemic brain injury in premature infants for which there is no effective treatment. The objective of this study was to determine whether transplanted mouse oligodendrocyte progenitor cells (OPCs) have neuroprotective effects in a rat model of PVL. Hypoxia-ischemia (HI) was induced in 3-day-old rat pups by left carotid artery ligation, followed by exposure to 6% oxygen for 2.5 h. Animals were assigned to OPC transplantation or sham control groups and injected with OPCs or PBS, respectively, and sacrificed up to 6 weeks later for immunohistochemical analysis to investigate the survival and differentiation of transplanted OPCs. Apoptosis was evaluated by double immunolabeling of brain sections for caspase-3 and neuronal nuclei (NeuN), while proliferation was assessed using a combination of anti-Nestin and -bromodeoxyuridine antibodies. The expression of brain-derived neurotrophic factor (BDNF) and Bcl-2 was examined 7 days after OPC transplantation. The Morris water maze was used to test spatial learning and memory. The results showed that transplanted OPCs survived and formed a myelin sheath, and stimulated BDNF and Bcl-2 expression and the proliferation of neural stem cells (NSC), while inhibiting HI-induced neuronal apoptosis relative to control animals. Moreover, deficits in spatial learning and memory resulting from HI were improved by OPC transplantation. These results demonstrate an important neuroprotective role for OPCs that can potentially be exploited in cell-based therapeutic approaches to minimize HI-induced brain injury.


Subject(s)
Brain Ischemia/therapy , Neural Stem Cells/transplantation , Oligodendroglia/metabolism , Stem Cell Transplantation , Allografts , Animals , Apoptosis , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain-Derived Neurotrophic Factor/biosynthesis , Caspase 3/biosynthesis , Gene Expression Regulation , Mice , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Oligodendroglia/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Rats
17.
Nat Commun ; 5: 5036, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25297674

ABSTRACT

Mutations in the X-linked gene encoding the transcriptional modulator methyl-CpG-binding protein 2 (MeCP2) impair postnatal development of the brain. Here we use neuronal-type specific gene deletion in mice to show that conditional Mecp2 deletion in GABAergic parvalbumin-expressing (PV) cells (PV-Mecp2(-/y)) does not cause most Rett-syndrome-like behaviours, but completely abolishes experience-dependent critical period plasticity of primary visual cortex (V1) that develops normal visual functions. However, selective loss of Mecp2 in GABAergic somatostatin-expressing cells or glutamatergic pyramidal cells does not affect the critical period plasticity. MeCP2-deficient PV cells exhibit high intrinsic excitability, selectively reduced efficacy of recurrent excitatory synapses in V1 layer 4 circuits, and decreased evoked visual responses in vivo. Enhancing cortical gamma-aminobutyric acid (GABA) inhibition with diazepam infusion can restore critical period plasticity in both young and adult PV-Mecp2(-/y) mice. Thus, MeCP2 expression in inhibitory PV cells during the critical period is essential for local circuit functions underlying experience-dependent cortical plasticity.


Subject(s)
Critical Period, Psychological , GABAergic Neurons/physiology , Methyl-CpG-Binding Protein 2/deficiency , Neuronal Plasticity/physiology , Visual Cortex/physiology , Animals , Crosses, Genetic , Diazepam/pharmacology , Flow Cytometry , GABAergic Neurons/metabolism , Gene Deletion , Immunohistochemistry , Male , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Neuronal Plasticity/drug effects , Neuronal Plasticity/genetics , Parvalbumins/metabolism , Patch-Clamp Techniques , Photic Stimulation , Real-Time Polymerase Chain Reaction , Visual Cortex/cytology
18.
Neurosci Bull ; 29(6): 773-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24136242

ABSTRACT

Autism is an etiologically heterogeneous group of neurodevelopmental disorders, diagnosed mostly by the clinical behavioral phenotypes. The concept that the tumor-related gene PTEN plays a critical role in autism spectrum disorder has emerged over the last decade. In this review, we focus on the essential role of the PTEN signaling pathway in neuronal differentiation and the formation of neural circuitry, as well as genetic mouse models with Pten manipulations. Particularly, accumulated data suggest that the effect of PTEN on neural stem-cell development contributes significantly to the pathophysiology of autism spectrum disorders.


Subject(s)
Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Signal Transduction , Animals , Disease Models, Animal , Humans , Mice , Neural Stem Cells/metabolism , Neurons/metabolism
19.
Article in English | MEDLINE | ID: mdl-12053204

ABSTRACT

About a third of the expressed genes had expression level dramatically changed during adipocyte differentiation. By locating their chromosomal genetic map positions, the positional correlation of these differentiation-regulated genes with their expression regulation was analyzed. The results indicated that there was no chromosomal positional effect in regulating these genes'expression during adipocyte differentiation.

SELECTION OF CITATIONS
SEARCH DETAIL
...