Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Elife ; 122023 10 13.
Article En | MEDLINE | ID: mdl-37830916

Dopamine system dysfunction is implicated in adolescent-onset neuropsychiatric disorders. Although psychosis symptoms can be alleviated by antipsychotics, cognitive symptoms remain unresponsive and novel paradigms investigating the circuit substrates underlying cognitive deficits are critically needed. The frontal cortex and its dopaminergic input from the midbrain are implicated in cognitive functions and undergo maturational changes during adolescence. Here, we used mice carrying mutations in Arc or Disc1 to model mesofrontal dopamine circuit deficiencies and test circuit-based neurostimulation strategies to restore cognitive functions. We found that in a memory-guided spatial navigation task, frontal cortical neurons were activated coordinately at the decision-making point in wild-type but not Arc-/- mice. Chemogenetic stimulation of midbrain dopamine neurons or optogenetic stimulation of frontal cortical dopamine axons in a limited adolescent period consistently reversed genetic defects in mesofrontal innervation, task-coordinated neuronal activity, and memory-guided decision-making at adulthood. Furthermore, adolescent stimulation of dopamine neurons also reversed the same cognitive deficits in Disc1+/- mice. Our findings reveal common mesofrontal circuit alterations underlying the cognitive deficits caused by two different genes and demonstrate the feasibility of adolescent neurostimulation to reverse these circuit and behavioral deficits. These results may suggest developmental windows and circuit targets for treating cognitive deficits in neurodevelopmental disorders.


Antipsychotic Agents , Dopamine , Animals , Mice , Dopamine/physiology , Frontal Lobe , Cognition , Prefrontal Cortex/physiology , Nerve Tissue Proteins
2.
Immunity ; 56(9): 2105-2120.e13, 2023 09 12.
Article En | MEDLINE | ID: mdl-37527657

Childhood neglect and/or abuse can induce mental health conditions with unknown mechanisms. Here, we identified stress hormones as strong inducers of astrocyte-mediated synapse phagocytosis. Using in vitro, in vivo, and human brain organoid experiments, we showed that stress hormones increased the expression of the Mertk phagocytic receptor in astrocytes through glucocorticoid receptor (GR). In post-natal mice, exposure to early social deprivation (ESD) specifically activated the GR-MERTK pathway in astrocytes, but not in microglia. The excitatory post-synaptic density in cortical regions was reduced in ESD mice, and there was an increase in the astrocytic engulfment of these synapses. The loss of excitatory synapses, abnormal neuronal network activities, and behavioral abnormalities in ESD mice were largely prevented by ablating GR or MERTK in astrocytes. Our work reveals the critical roles of astrocytic GR-MERTK activation in evoking stress-induced abnormal behaviors in mice, suggesting GR-MERTK signaling as a therapeutic target for stress-induced mental health conditions.


Astrocytes , Phagocytosis , Stress, Psychological , Animals , Child , Humans , Mice , Astrocytes/metabolism , c-Mer Tyrosine Kinase/genetics , Hormones/metabolism , Synapses/metabolism , Stress, Psychological/metabolism
3.
Stem Cell Reports ; 18(7): 1468-1481, 2023 07 11.
Article En | MEDLINE | ID: mdl-37390823

Quiescence is a hallmark of adult neural stem cells (NSCs) in the mammalian brain, and establishment and maintenance of quiescence is essential for life-long continuous neurogenesis. How NSCs in the dentate gyrus (DG) of the hippocampus acquire their quiescence during early postnatal stages and continuously maintain quiescence in adulthood is poorly understood. Here, we show that Hopx-CreERT2-mediated conditional deletion of Nkcc1, which encodes a chloride importer, in mouse DG NSCs impairs both their quiescence acquisition at early postnatal stages and quiescence maintenance in adulthood. Furthermore, PV-CreERT2-mediated deletion of Nkcc1 in PV interneurons in the adult mouse brain leads to activation of quiescent DG NSCs, resulting in an expanded NSC pool. Consistently, pharmacological inhibition of NKCC1 promotes NSC proliferation in both early postnatal and adult mouse DG. Together, our study reveals both cell-autonomous and non-cell-autonomous roles of NKCC1 in regulating the acquisition and maintenance of NSC quiescence in the mammalian hippocampus.


Hippocampus , Neural Stem Cells , Animals , Mice , Neurogenesis/physiology , Cell Division , Dentate Gyrus , Mammals
4.
bioRxiv ; 2023 Jul 12.
Article En | MEDLINE | ID: mdl-36778456

Dopamine system dysfunction is commonly implicated in adolescent-onset neuropsychiatric disorders. Although psychosis symptoms can be alleviated by antipsychotics, cognitive symptoms remain unresponsive to such pharmacological treatments and novel research paradigms investigating the circuit substrates underlying cognitive deficits are critically needed. The frontal cortex and its dopaminergic input from the midbrain are implicated in cognitive functions and undergo maturational changes during adolescence. Here, we used mice carrying mutations in the Arc or DISC1 genes to model mesofrontal dopamine circuit deficiencies and test circuit-based neurostimulation strategies to restore cognitive functions. We found that in a memory-guided spatial navigation task, frontal cortical neurons were activated coordinately at the decision-making point in wild-type but not Arc mutant mice. Chemogenetic stimulation of midbrain dopamine neurons or optogenetic stimulation of frontal cortical dopamine axons in a limited adolescent period consistently reversed genetic defects in mesofrontal innervation, task-coordinated neuronal activity, and memory-guided decision-making at adulthood. Furthermore, adolescent stimulation of dopamine neurons also reversed the same cognitive deficits in DISC1 mutant mice. Our findings reveal common mesofrontal circuit alterations underlying the cognitive deficits caused by two different genes and demonstrate the feasibility of adolescent neurostimulation to reverse these circuit and behavioral deficits. These results may suggest developmental windows and circuit targets for treating cognitive deficits in neurodevelopmental disorders.

5.
Cell Stem Cell ; 30(3): 300-311.e11, 2023 03 02.
Article En | MEDLINE | ID: mdl-36764294

Increasing evidence implicates the critical roles of various epitranscriptomic RNA modifications in different biological processes. Methyltransferase METTL8 installs 3-methylcytosine (m3C) modification of mitochondrial tRNAs in vitro; however, its role in intact biological systems is unknown. Here, we show that Mettl8 is localized in mitochondria and installs m3C specifically on mitochondrial tRNAThr/Ser(UCN) in mouse embryonic cortical neural stem cells. At molecular and cellular levels, Mettl8 deletion in cortical neural stem cells leads to reduced mitochondrial protein translation and attenuated respiration activity. At the functional level, conditional Mettl8 deletion in mice results in impaired embryonic cortical neural stem cell maintenance in vivo, which can be rescued by pharmacologically enhancing mitochondrial functions. Similarly, METTL8 promotes mitochondrial protein expression and neural stem cell maintenance in human forebrain cortical organoids. Together, our study reveals a conserved epitranscriptomic mechanism of Mettl8 and mitochondrial tRNA m3C modification in maintaining embryonic cortical neural stem cells in mice and humans.


Methyltransferases , Mitochondria , Mice , Animals , Humans , Mitochondria/metabolism , Methyltransferases/genetics , RNA, Transfer/metabolism , Neurogenesis , Mitochondrial Proteins/metabolism
6.
Neurosci Res ; 187: 3-13, 2023 Feb.
Article En | MEDLINE | ID: mdl-36170922

Based on experience during our life, neuronal connectivity continuously changes through structural remodeling of synapses. Recent studies have shown that the complex interaction between astrocytes and synapses regulates structural synapse remodeling by inducing the formation and elimination of synapses, as well as their functional maturation. Defects in this astrocyte-mediated synapse remodeling cause problems in not only neuronal network activities but also animal behaviors. Moreover, in various neurological disorders, astrocytes have been shown to play central roles in the initiation and progression of synaptic pathophysiology through impaired interactions with synapses. In this review, we will discuss recent studies identifying the novel roles of astrocytes in neuronal circuit remodeling, focusing on synapse formation and elimination. We will also discuss the potential implication of defective astrocytic function in evoking various brain disorders.


Brain Diseases , Nervous System Diseases , Animals , Astrocytes/physiology , Synapses/physiology , Neurons/physiology , Neuronal Plasticity/physiology
7.
Biol Psychiatry ; 92(10): 815-826, 2022 11 15.
Article En | MEDLINE | ID: mdl-34247782

BACKGROUND: Gene dosage imbalance caused by copy number variations (CNVs) is a prominent contributor to brain disorders. In particular, 15q11.2 CNV duplications and deletions have been associated with autism spectrum disorder and schizophrenia, respectively. The mechanism underlying these diametric contributions remains unclear. METHODS: We established both loss-of-function and gain-of-function mouse models of Cyfip1, one of four genes within 15q11.2 CNVs. To assess the functional consequences of altered CYFIP1 levels, we performed systematic investigations on behavioral, electrophysiological, and biochemical phenotypes in both mouse models. In addition, we utilized RNA immunoprecipitation sequencing (RIP-seq) analysis to reveal molecular targets of CYFIP1 in vivo. RESULTS: Cyfip1 loss-of-function and gain-of function mouse models exhibited distinct and shared behavioral abnormalities related to autism spectrum disorder and schizophrenia. RIP-seq analysis identified messenger RNA targets of CYFIP1 in vivo, including postsynaptic NMDA receptor (NMDAR) complex components. In addition, these mouse models showed diametric changes in levels of postsynaptic NMDAR complex components at synapses because of dysregulated protein translation, resulting in bidirectional alteration of NMDAR-mediated signaling. Importantly, pharmacological balancing of NMDAR signaling in these mouse models with diametric Cyfip1 dosages rescues behavioral abnormalities. CONCLUSIONS: CYFIP1 regulates protein translation of NMDAR and associated complex components at synapses to maintain normal synaptic functions and behaviors. Our integrated analyses provide insight into how gene dosage imbalance caused by CNVs may contribute to divergent neuropsychiatric disorders.


Autism Spectrum Disorder , Mental Disorders , Mice , Animals , Receptors, N-Methyl-D-Aspartate/genetics , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , DNA Copy Number Variations , Mice, Inbred C57BL , N-Methylaspartate/genetics , Adaptor Proteins, Signal Transducing/genetics , Disease Models, Animal , RNA, Messenger , RNA
8.
Neurobiol Stress ; 15: 100373, 2021 Nov.
Article En | MEDLINE | ID: mdl-34401409

Depression is accompanied by neuronal atrophy and decreased neuroplasticity. Leucine-rich glioma-inactivated protein 1 (LGI1), a metastasis suppressor, plays an important role in the development of CNS synapses. We found that LGI1 expression was reduced in the hippocampi of mice that underwent chronic unpredictable stress (CUS), and could be rescued by the antidepressant, fluoxetine. Recombinant soluble neuritin, an endogenous protein previously implicated in antidepressant-like behaviors, elevated hippocampal LGI1 expression in a manner dependent on histone deacetylase 5 (HDAC5) phosphorylation. Accordingly, Nrn1 flox/flox ;Pomc-cre (Nrn1 cOE) mice, which conditionally overexpress neuritin, displayed increases in hippocampal LGI1 level under CUS and exhibited resilience to CUS that were blocked by hippocampal depletion of LGI1. Interestingly, neuritin-mediated LGI1 expression was inhibited by HNMPA-(AM)3, an insulin receptor inhibitor, as was neuritin-mediated HDAC5 phosphorylation. We thus establish hippocampal LGI1 as an effector of neurite outgrowth and stress resilience, and suggest that HDAC5-LGI1 plays a critical role in ameliorating pathological depression.

10.
Nat Commun ; 12(1): 1398, 2021 03 03.
Article En | MEDLINE | ID: mdl-33658519

We previously identified a causal link between a rare patient mutation in DISC1 (disrupted-in-schizophrenia 1) and synaptic deficits in cortical neurons differentiated from isogenic patient-derived induced pluripotent stem cells (iPSCs). Here we find that transcripts related to phosphodiesterase 4 (PDE4) signaling are significantly elevated in human cortical neurons differentiated from iPSCs with the DISC1 mutation and that inhibition of PDE4 or activation of the cAMP signaling pathway functionally rescues synaptic deficits. We further generated a knock-in mouse line harboring the same patient mutation in the Disc1 gene. Heterozygous Disc1 mutant mice exhibit elevated levels of PDE4s and synaptic abnormalities in the brain, and social and cognitive behavioral deficits. Pharmacological inhibition of the PDE4 signaling pathway rescues these synaptic, social and cognitive behavioral abnormalities. Our study shows that patient-derived isogenic iPSC and humanized mouse disease models are integral and complementary for translational studies with a better understanding of underlying molecular mechanisms.


Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Induced Pluripotent Stem Cells/drug effects , Nerve Tissue Proteins/genetics , Phosphodiesterase 4 Inhibitors/pharmacology , Schizophrenia/genetics , Animals , Behavior, Animal/drug effects , Cerebral Cortex/physiology , Disease Models, Animal , Female , Gene Expression , Humans , Male , Mice, Mutant Strains , Mutation , Neurons/drug effects , Rolipram/pharmacology , Schizophrenia/pathology , Synapses/drug effects , Synapses/physiology
11.
J Neurosci ; 40(10): 2015-2024, 2020 03 04.
Article En | MEDLINE | ID: mdl-31988061

Neural stem cells (NSCs) persist throughout life in the subventricular zone (SVZ) neurogenic niche of the lateral ventricles as Type B1 cells in adult mice. Maintaining this population of NSCs depends on the balance between quiescence and self-renewing or self-depleting cell divisions. Interactions between B1 cells and the surrounding niche are important in regulating this balance, but the mechanisms governing these processes have not been fully elucidated. The cytoplasmic FMRP-interacting protein (Cyfip1) regulates apical-basal polarity in the embryonic brain. Loss of Cyfip1 during embryonic development in mice disrupts the embryonic niche and affects cortical neurogenesis. However, a direct role for Cyfip1 in the regulation of adult NSCs has not been established. Here, we demonstrate that Cyfip1 expression is preferentially localized to B1 cells in the adult mouse SVZ. Loss of Cyfip1 in the embryonic mouse brain results in altered adult SVZ architecture and expansion of the adult B1 cell population at the ventricular surface. Furthermore, acute deletion of Cyfip1 in adult NSCs results in a rapid change in adherens junction proteins as well as increased proliferation and number of B1 cells at the ventricular surface. Together, these data indicate that Cyfip1 plays a critical role in the formation and maintenance of the adult SVZ niche; furthermore, deletion of Cyfip1 unleashes the capacity of adult B1 cells for symmetric renewal to increase the adult NSC pool.SIGNIFICANCE STATEMENT Neural stem cells (NSCs) persist in the subventricular zone of the lateral ventricles in adult mammals, and the size of this population is determined by the balance between quiescence and self-depleting or renewing cell division. The mechanisms regulating these processes are not fully understood. This study establishes that the cytoplasmic FMRP interacting protein 1 (Cyfip1) regulates NSC fate decisions in the adult subventricular zone and adult NSCs that are quiescent or typically undergo self-depleting divisions retain the ability to self-renew. These results contribute to our understanding of how adult NSCs are regulated throughout life and has potential implications for human brain disorders.


Adaptor Proteins, Signal Transducing/metabolism , Lateral Ventricles/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Stem Cell Niche/physiology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Aging , Animals , Lateral Ventricles/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/cytology
12.
Development ; 145(14)2018 07 25.
Article En | MEDLINE | ID: mdl-29950389

Mammary glands develop through primary ductal elongation and side branching to maximize the spatial area. Although primary ducts are generated by bifurcation of terminal end buds, the mechanism through which side branching occurs is still largely unclear. Here, we show that inhibitor of DNA-binding 2 (ID2) drives side branch formation through the differentiation of K6+ bipotent progenitor cells (BPs) into CD61+ luminal progenitor cells (LPs). Id2-null mice had side-branching defects, along with developmental blockage of the differentiation of K6+ BPs into CD61+ LPs. Notably, CD61+ LPs were found in budding and side branches, but not in terminal end buds. Hormone reconstitution studies using ovariectomized MMTV-hemagglutinin-nuclear localized sequence-tagged Id2 transgenic mice revealed that ID2 is a key mediator of progesterone, which drives luminal lineage differentiation and side branching. Our results suggest that CD61 is a marker of side branches and that ID2 regulates side branch formation by inducing luminal lineage commitment from K6+ BPs to CD61+ LPs.


Body Patterning , Cell Lineage , Inhibitor of Differentiation Protein 2/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/embryology , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Differentiation , Cell Nucleus/metabolism , Female , Gene Deletion , Imaging, Three-Dimensional , Integrin beta3/metabolism , Mice , Models, Biological , Progesterone/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
13.
Cell ; 171(4): 877-889.e17, 2017 Nov 02.
Article En | MEDLINE | ID: mdl-28965759

N6-methyladenosine (m6A), installed by the Mettl3/Mettl14 methyltransferase complex, is the most prevalent internal mRNA modification. Whether m6A regulates mammalian brain development is unknown. Here, we show that m6A depletion by Mettl14 knockout in embryonic mouse brains prolongs the cell cycle of radial glia cells and extends cortical neurogenesis into postnatal stages. m6A depletion by Mettl3 knockdown also leads to a prolonged cell cycle and maintenance of radial glia cells. m6A sequencing of embryonic mouse cortex reveals enrichment of mRNAs related to transcription factors, neurogenesis, the cell cycle, and neuronal differentiation, and m6A tagging promotes their decay. Further analysis uncovers previously unappreciated transcriptional prepatterning in cortical neural stem cells. m6A signaling also regulates human cortical neurogenesis in forebrain organoids. Comparison of m6A-mRNA landscapes between mouse and human cortical neurogenesis reveals enrichment of human-specific m6A tagging of transcripts related to brain-disorder risk genes. Our study identifies an epitranscriptomic mechanism in heightened transcriptional coordination during mammalian cortical neurogenesis.


Neurogenesis , Prosencephalon/embryology , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Animals , Cell Cycle , Gene Expression Regulation , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Mice , Mice, Knockout , Neural Stem Cells/metabolism , Organoids/metabolism , Prosencephalon/cytology , Prosencephalon/metabolism , RNA Stability
14.
Cell Stem Cell ; 21(3): 349-358.e6, 2017 09 07.
Article En | MEDLINE | ID: mdl-28826723

Zika virus (ZIKV) directly infects neural progenitors and impairs their proliferation. How ZIKV interacts with the host molecular machinery to impact neurogenesis in vivo is not well understood. Here, by systematically introducing individual proteins encoded by ZIKV into the embryonic mouse cortex, we show that expression of ZIKV-NS2A, but not Dengue virus (DENV)-NS2A, leads to reduced proliferation and premature differentiation of radial glial cells and aberrant positioning of newborn neurons. Mechanistically, in vitro mapping of protein-interactomes and biochemical analysis suggest interactions between ZIKA-NS2A and multiple adherens junction complex (AJ) components. Functionally, ZIKV-NS2A, but not DENV-NS2A, destabilizes the AJ complex, resulting in impaired AJ formation and aberrant radial glial fiber scaffolding in the embryonic mouse cortex. Similarly, ZIKA-NS2A, but not DENV-NS2A, reduces radial glial cell proliferation and causes AJ deficits in human forebrain organoids. Together, our results reveal pathogenic mechanisms underlying ZIKV infection in the developing mammalian brain.


Adherens Junctions/metabolism , Cerebral Cortex/metabolism , Mammals/metabolism , Membrane Proteins/metabolism , Neurogenesis , Proteolysis , Viral Nonstructural Proteins/metabolism , Zika Virus/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cerebral Cortex/embryology , HEK293 Cells , Humans , Mice , Neuroglia/pathology , Protein Binding , Protein Interaction Mapping , Zika Virus Infection/pathology
15.
Nature ; 515(7527): 414-8, 2014 Nov 20.
Article En | MEDLINE | ID: mdl-25132547

Dysregulated neurodevelopment with altered structural and functional connectivity is believed to underlie many neuropsychiatric disorders, and 'a disease of synapses' is the major hypothesis for the biological basis of schizophrenia. Although this hypothesis has gained indirect support from human post-mortem brain analyses and genetic studies, little is known about the pathophysiology of synapses in patient neurons and how susceptibility genes for mental disorders could lead to synaptic deficits in humans. Genetics of most psychiatric disorders are extremely complex due to multiple susceptibility variants with low penetrance and variable phenotypes. Rare, multiply affected, large families in which a single genetic locus is probably responsible for conferring susceptibility have proven invaluable for the study of complex disorders. Here we generated induced pluripotent stem (iPS) cells from four members of a family in which a frameshift mutation of disrupted in schizophrenia 1 (DISC1) co-segregated with major psychiatric disorders and we further produced different isogenic iPS cell lines via gene editing. We showed that mutant DISC1 causes synaptic vesicle release deficits in iPS-cell-derived forebrain neurons. Mutant DISC1 depletes wild-type DISC1 protein and, furthermore, dysregulates expression of many genes related to synapses and psychiatric disorders in human forebrain neurons. Our study reveals that a psychiatric disorder relevant mutation causes synapse deficits and transcriptional dysregulation in human neurons and our findings provide new insight into the molecular and synaptic etiopathology of psychiatric disorders.


Induced Pluripotent Stem Cells/pathology , Mental Disorders/pathology , Synapses/pathology , Animals , Cell Differentiation , Fibroblasts , Glutamine/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mental Disorders/genetics , Mental Disorders/metabolism , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Pedigree , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Prosencephalon/metabolism , Prosencephalon/pathology , Protein Binding , Synapses/metabolism , Transcriptome
16.
Cell Stem Cell ; 15(1): 79-91, 2014 Jul 03.
Article En | MEDLINE | ID: mdl-24996170

Defects in brain development are believed to contribute toward the onset of neuropsychiatric disorders, but identifying specific underlying mechanisms has proven difficult. Here, we took a multifaceted approach to investigate why 15q11.2 copy number variants are prominent risk factors for schizophrenia and autism. First, we show that human iPSC-derived neural progenitors carrying 15q11.2 microdeletion exhibit deficits in adherens junctions and apical polarity. This results from haploinsufficiency of CYFIP1, a gene within 15q11.2 that encodes a subunit of the WAVE complex, which regulates cytoskeletal dynamics. In developing mouse cortex, deficiency in CYFIP1 and WAVE signaling similarly affects radial glial cells, leading to their ectopic localization outside of the ventricular zone. Finally, targeted human genetic association analyses revealed an epistatic interaction between CYFIP1 and WAVE signaling mediator ACTR2 and risk for schizophrenia. Our findings provide insight into how CYFIP1 regulates neural stem cell function and may contribute to the susceptibility of neuropsychiatric disorders.


Adherens Junctions/genetics , Autistic Disorder/metabolism , Induced Pluripotent Stem Cells/physiology , Neural Stem Cells/physiology , Schizophrenia/metabolism , Actin-Related Protein 2/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adherens Junctions/pathology , Adult , Animals , Autistic Disorder/genetics , Autistic Disorder/pathology , Cell Line , Cell Polarity/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 15/genetics , DNA Copy Number Variations , Epistasis, Genetic , Genetic Association Studies , Haploinsufficiency , Humans , Intellectual Disability/genetics , Male , Mice , Mice, Inbred Strains , Middle Aged , Risk , Schizophrenia/genetics , Schizophrenia/pathology , White People , Wiskott-Aldrich Syndrome Protein Family/metabolism
17.
Mol Brain ; 5: 40, 2012 Oct 30.
Article En | MEDLINE | ID: mdl-23111145

BACKGROUND: Notch signaling is well recognized as a key regulator of the neuronal fate during embryonic development, but its function in the adult brain is still largely unknown. Mind bomb-1 (Mib1) is an essential positive regulator in the Notch pathway, acting non-autonomously in the signal-sending cells. Therefore, genetic ablation of Mib1 in mature neuron would give valuable insight to understand the cell-to-cell interaction between neurons via Notch signaling for their proper function. RESULTS: Here we show that the inactivation of Mib1 in mature neurons in forebrain results in impaired hippocampal dependent spatial memory and contextual fear memory. Consistently, hippocampal slices from Mib1-deficient mice show impaired late-phase, but not early-phase, long-term potentiation and long-term depression without change in basal synaptic transmission at SC-CA1 synapses. CONCLUSIONS: These data suggest that Mib1-mediated Notch signaling is essential for long-lasting synaptic plasticity and memory formation in the rodent hippocampus.


Memory, Long-Term/physiology , Neuronal Plasticity/physiology , Receptors, Notch/metabolism , Signal Transduction , Synapses/physiology , Ubiquitin-Protein Ligases/metabolism , Aging/metabolism , Animals , Hippocampus/anatomy & histology , Hippocampus/enzymology , Long-Term Potentiation , Mice , Mice, Knockout , Neurons/metabolism , Phenotype , Protein Kinase C/metabolism , Protein Structure, Tertiary , Receptors, Notch/chemistry
18.
Mol Cell Biol ; 31(23): 4775-88, 2011 Dec.
Article En | MEDLINE | ID: mdl-21947283

RANKL plays an essential role in mammary gland development during pregnancy. However, the molecular mechanism by which RANK signaling leads to mammary gland development is largely unknown. We report here that RANKL stimulation induces phosphorylation of Id2 at serine 5, which leads to nuclear retention of Id2. In lactating Id2Tg; RANKL(-/-) mice, Id2 was not phosphorylated and was localized in the cytoplasm. In addition, in lactating Id2(S5A)Tg mice, Id2(S5A) (with serine 5 mutated to alanine) was exclusively localized in the cytoplasm of mammary epithelial cells (MECs), while endogenous Id2 was localized in the nucleus. Intriguingly, nuclear expression of Id2(S5A) rescued increased apoptosis and defective differentiation of MECs in RANKL(-/-) mice. Our results demonstrate that nuclear retention of Id2 due to RANK signaling plays a decisive role in the survival and differentiation of MECs during mammary gland development.


Cell Differentiation , Cell Nucleus/metabolism , Cell Survival , Epithelial Cells/physiology , Inhibitor of Differentiation Protein 2/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Receptor Activator of Nuclear Factor-kappa B/metabolism , Animals , Cell Line, Tumor , Epithelial Cells/metabolism , Female , Gene Expression , Gene Knockout Techniques , Humans , Inhibitor of Differentiation Protein 2/genetics , Lactation , Male , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence , Milk Proteins/genetics , Milk Proteins/metabolism , Phosphorylation , Pregnancy , Protein Transport , Receptor Activator of Nuclear Factor-kappa B/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction
19.
Ann Allergy Asthma Immunol ; 104(4): 343-8, 2010 Apr.
Article En | MEDLINE | ID: mdl-20408346

BACKGROUND: Probiotics have been suggested to be useful in children with atopic eczema-dermatitis syndrome (AEDS). OBJECTIVE: To assess the clinical effect of Lactobacillus sakei supplementation in children with AEDS. METHODS: In a double-blind, placebo-controlled trial, children aged 2 to 10 years with AEDS with a minimum SCORing of Atopic Dermatitis (SCORAD) score of 25 were randomized to receive either daily L sakei KCTC 10755BP or daily placebo supplementation for 12 weeks. Changes in SCORAD scores and serum chemokine levels from baseline were evaluated. RESULTS: Eighty-eight children were enrolled, and 45 were allocated to probiotic treatment. Seventy-five children completed the study, with 4 dropouts in the probiotic group and 9 in the placebo group. At week 12, SCORAD total scores adjusted by pretreatment values were lower after probiotic treatment than after placebo treatment (P = .01). There was a 31% (13.1-point) improvement in mean disease activity with probiotic use compared with a 13% (5.2-point) improvement with placebo use (P = .008). Significant differences in favor of probiotic treatment were also observed in proportions of patients achieving improvement of at least 30% and 50%. Compared with placebo, probiotic administration was associated with lower pretreatment-adjusted serum levels of CCL17 and CCL27 (P =.03 for both), which were significantly correlated with SCORAD total score (r = 0.59 and 0.63, respectively; P < .001). CONCLUSIONS: Supplementation of L sakei in children with AEDS was associated with a substantial clinical improvement and a significant decrease in chemokine levels, reflecting the severity of AEDS.


Dermatitis, Atopic/therapy , Lactobacillus , Probiotics/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Chemokine CCL17/blood , Chemokine CCL27/blood , Child , Child, Preschool , Dermatitis, Atopic/blood , Dermatitis, Atopic/diagnosis , Double-Blind Method , Female , Humans , Male , Severity of Illness Index , Treatment Outcome
20.
J Biol Chem ; 284(48): 33634-41, 2009 Nov 27.
Article En | MEDLINE | ID: mdl-19801644

Although terminal differentiation of intestinal epithelium is essential for the efficient digestion and absorption of nutrients, little is known about the molecular mechanisms underlying this process. Recent studies have shown that Elf3 (E74-like factor 3), a member of the ETS transcription factor family, has an essential role in the terminal differentiation of absorptive enterocytes and mucus-secreting goblet cells. Here, we demonstrated that Crif1 (CR6-interacting factor 1) functions as transcriptional coactivator of Elf3 in intestinal epithelium differentiation. The intestinal epithelium-specific Crif1-deficient mice died soon after birth and displayed severe alterations of tissue architecture in the small intestine, including poor microvillus formation and abnormal differentiation of absorptive enterocytes. Strikingly, these phenotypes are largely similar to that of Elf3-deficient mice, suggesting that Elf3 signaling in the intestinal epithelium depends on the Crif1 expression. We dissected this relationship further and found that Crif1 indeed interacted with Elf3 through its ETS DNA binding domain and enhanced the transcriptional activity of Elf3 by regulating the DNA binding activity. Knockdown of Crif1 by RNA interference conversely attenuated the transcriptional activity of Elf3. Consistently, the expression level of Tgf-betaRII (transforming growth factor beta type II receptor), a critical target gene of Elf3, was dramatically reduced in the Crif1-deficient mice. Our results reveal that Crif1 is a novel and essential transcriptional coactivator of Elf3 for the terminal differentiation of absorptive enterocytes.


Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Intestinal Mucosa/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , COS Cells , Cell Cycle Proteins/genetics , Cell Differentiation , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , DNA-Binding Proteins/genetics , Enterocytes/metabolism , Enterocytes/pathology , Epithelium/embryology , Epithelium/metabolism , Epithelium/pathology , Female , Gene Expression Regulation, Developmental , HCT116 Cells , Humans , In Situ Hybridization , Intestines/embryology , Intestines/pathology , Male , Mice , Mice, Knockout , Mutation , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transcription Factors/genetics , Transfection
...