Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Mol Psychiatry ; 26(12): 7550-7559, 2021 12.
Article in English | MEDLINE | ID: mdl-34262135

ABSTRACT

Recent evidence has documented the potential roles of histone-modifying enzymes in autism-spectrum disorder (ASD). Aberrant histone H3 lysine 9 (H3K9) dimethylation resulting from genetic variants in histone methyltransferases is known for neurodevelopmental and behavioral anomalies. However, a systematic examination of H3K9 methylation dynamics in ASD is lacking. Here we resequenced nine genes for histone methyltransferases and demethylases involved in H3K9 methylation in individuals with ASD and healthy controls using targeted next-generation sequencing. We identified a novel rare variant (A211S) in the SUV39H2, which was predicted to be deleterious. The variant showed strongly reduced histone methyltransferase activity in vitro. In silico analysis showed that the variant destabilizes the hydrophobic core and allosterically affects the enzyme activity. The Suv39h2-KO mice displayed hyperactivity and reduced behavioral flexibility in learning the tasks that required complex behavioral adaptation, which is relevant for ASD. The Suv39h2 deficit evoked an elevated expression of a subset of protocadherin ß (Pcdhb) cluster genes in the embryonic brain, which is attributable to the loss of H3K9 trimethylation (me3) at the gene promoters. Reduced H3K9me3 persisted in the cerebellum of Suv39h2-deficient mice to an adult stage. Congruently, reduced expression of SUV39H1 and SUV39H2 in the postmortem brain samples of ASD individuals was observed, underscoring the role of H3K9me3 deficiency in ASD etiology. The present study provides direct evidence for the role of SUV39H2 in ASD and suggests a molecular cascade of SUV39H2 dysfunction leading to H3K9me3 deficiency followed by an untimely, elevated expression of Pcdhb cluster genes during early neurodevelopment.


Subject(s)
Autistic Disorder , Histone-Lysine N-Methyltransferase/genetics , Animals , Brain/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Mice , Protocadherins
2.
Cereb Cortex ; 31(1): 448-462, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32924060

ABSTRACT

Structural changes in the corpus callosum have been reported in schizophrenia; however, the underlying molecular mechanism remains unclear. As the corpus callosum is high in lipid content, we analyzed the lipid contents of the corpora callosa from 15 patients with schizophrenia and 15 age- and sex-matched controls using liquid chromatography coupled to tandem mass spectrometry and identified lipid combinations associated with schizophrenia. Real-time quantitative polymerase chain reaction analyses using extended samples (schizophrenia, n = 95; control, n = 91) showed low expression levels of lipid metabolism-related genes and their potential upstream transcription factors in schizophrenia. Subsequent pathway analysis identified a gene regulatory network where nuclear factor of activated T cells 2 (NFATC2) is placed most upstream. We also observed low gene expression levels of microglial markers, inflammatory cytokines, and colony-stimulating factor 1 receptor (CSF1R), which is known to regulate the density of microglia, in the corpus callosum in schizophrenia. The interactions between CSF1R and several genes in the presently identified gene network originating from NFATC2 have been reported. Collectively, this study provides evidence regarding lipid abnormalities in the corpora callosa of patients with schizophrenia and proposes the potential role of impaired "NFATC2-relevant gene network-microglial axis" as its underlying mechanism.


Subject(s)
Biomarkers/analysis , Corpus Callosum/pathology , Lipids , Microglia/pathology , Schizophrenia/pathology , Adult , Chromatography, Liquid/methods , Corpus Callosum/metabolism , Cytokines/metabolism , Female , Gene Regulatory Networks/physiology , Humans , Male , Microglia/metabolism , Middle Aged , Schizophrenia/metabolism
3.
Biosci Biotechnol Biochem ; 83(1): 114-122, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30200826

ABSTRACT

The present study describes the hair growth-promoting effects of sodium thiosulfate (STS), a widely used compound, in mice. STS accelerated hair growth in the "telogen model", suggesting that it stimulates telogen hair follicles to reenter the anagen phase of hair growth. In the same model, STS potentiated hair growth in an additive manner with minoxidil (MXD), a drug used for the treatment of androgenic alopecia. Furthermore, in the "anagen model", STS promoted hair growth, probably by promoting hair follicle proliferation. Since STS elevated the skin surface temperature, its hair growth-promoting activity may be partly due to vasorelaxation, similar to MXD. In addition, STS is known to generate a gaseous mediator, H2S, which has vasorelaxation and anti-inflammatory/anti-oxidative stress activities. Therefore, STS and/or provisionally its metabolite, H2S, may aid the hair growth process. Collectively, these results suggest that salts of thiosulfate may represent a novel and beneficial remedy for hair loss.


Subject(s)
Hair Follicle/drug effects , Hair Follicle/growth & development , Models, Animal , Thiosulfates/pharmacology , Alopecia/drug therapy , Animals , Drug Synergism , Gene Expression Regulation/drug effects , Humans , Male , Mice, Inbred C3H , Minoxidil/administration & dosage , Minoxidil/adverse effects , Minoxidil/pharmacology , Models, Biological , Skin Temperature/drug effects , Sulfurtransferases/genetics , Sulfurtransferases/metabolism , Thiosulfates/administration & dosage , Thiosulfates/adverse effects
4.
Glia ; 64(1): 48-62, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26296243

ABSTRACT

Fatty acid binding protein 7 (FABP7) expressed by astrocytes in developing and mature brains is involved in uptake and transportation of fatty acids, signal transduction, and gene transcription. Fabp7 knockout (Fabp7 KO) mice show behavioral phenotypes reminiscent of human neuropsychiatric disorders such as schizophrenia. However, direct evidence showing how FABP7 deficiency in astrocytes leads to altered brain function is lacking. Here, we examined neuronal dendritic morphology and synaptic plasticity in medial prefrontal cortex (mPFC) of Fabp7 KO mice and in primary cortical neuronal cultures. Golgi staining of cortical pyramidal neurons in Fabp7 KO mice revealed aberrant dendritic morphology and decreased spine density compared with those in wild-type (WT) mice. Aberrant dendritic morphology was also observed in primary cortical neurons co-cultured with FABP7-deficient astrocytes and neurons cultured in Fabp7 KO astrocyte-conditioned medium. Excitatory synapse number was decreased in mPFC of Fabp7 KO mice and in neurons co-cultured with Fabp7 KO astrocytes. Accordingly, whole-cell voltage-clamp recording in brain slices from pyramidal cells in the mPFC showed that both amplitude and frequency of action potential-independent miniature excitatory postsynaptic currents (mEPSCs) were decreased in Fabp7 KO mice. Moreover, transplantation of WT astrocytes into the mPFC of Fabp7 KO mice partially attenuated behavioral impairments. Collectively, these results suggest that astrocytic FABP7 is important for dendritic arbor growth, neuronal excitatory synapse formation, and synaptic transmission, and provide new insights linking FABP7, lipid homeostasis, and neuropsychiatric disorders, leading to novel therapeutic interventions.


Subject(s)
Astrocytes/physiology , Dendrites/physiology , Fatty Acid-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Prefrontal Cortex/physiology , Pyramidal Cells/physiology , Synapses/physiology , Animals , Astrocytes/transplantation , Coculture Techniques , Excitatory Postsynaptic Potentials/physiology , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Miniature Postsynaptic Potentials/physiology , Motor Activity/physiology , Nerve Tissue Proteins/genetics , Prefrontal Cortex/cytology , Prefrontal Cortex/surgery , Pyramidal Cells/cytology
5.
Hum Mol Genet ; 23(24): 6495-511, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25027319

ABSTRACT

Disturbances of lipid metabolism have been implicated in psychiatric illnesses. We previously reported an association between the gene for fatty acid binding protein 7 (FABP7) and schizophrenia. Furthermore, we identified and reported several rare non-synonymous polymorphisms of the brain-expressed genes FABP3, FABP5 and FABP7 from schizophrenia and autism spectrum disorder (ASD), diseases known to part share genetic architecture. Here, we conducted further studies to better understand the contribution these genes make to the pathogenesis of schizophrenia and ASD. In postmortem brains, we detected altered mRNA expression levels of FABP5 in schizophrenia, and of FABP7 in ASD and altered FABP5 in peripheral lymphocytes. Using a patient cohort, comprehensive mutation screening identified six missense and two frameshift variants from the three FABP genes. The two frameshift proteins, FABP3 E132fs and FABP7 N80fs, formed cellular aggregates and were unstable when expressed in cultured cells. The four missense mutants with predicted possible damaging outcomes showed no changes in intracellular localization. Examining ligand binding properties, FABP7 S86G and FABP7 V126L lost their preference for docosahexaenoic acid to linoleic acid. Finally, mice deficient in Fabp3, Fabp5 and Fabp7 were evaluated in a systematic behavioral test battery. The Fabp3 knockout (KO) mice showed decreased social memory and novelty seeking, and Fabp7 KO mice displayed hyperactive and anxiety-related phenotypes, while Fabp5 KO mice showed no apparent phenotypes. In conclusion, disturbances in brain-expressed FABPs could represent an underlying disease mechanism in a proportion of schizophrenia and ASD sufferers.


Subject(s)
Behavior, Animal , Carrier Proteins/genetics , Child Development Disorders, Pervasive/genetics , Fatty Acid-Binding Proteins/genetics , Schizophrenia/genetics , Tumor Suppressor Proteins/genetics , Amino Acid Sequence , Animals , Anxiety/genetics , Anxiety/physiopathology , Brain/metabolism , Brain/pathology , Carrier Proteins/metabolism , Child Development Disorders, Pervasive/metabolism , Child Development Disorders, Pervasive/physiopathology , Docosahexaenoic Acids/metabolism , Exploratory Behavior , Fatty Acid Binding Protein 3 , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins/metabolism , Frameshift Mutation , Humans , Linoleic Acid/metabolism , Lymphocytes/metabolism , Mice , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Schizophrenia/metabolism , Schizophrenia/physiopathology , Sequence Alignment , Tumor Suppressor Proteins/metabolism
6.
Glia ; 63(5): 780-94, 2015 May.
Article in English | MEDLINE | ID: mdl-25601031

ABSTRACT

Fatty acid-binding proteins (FABPs) bind and solubilize long-chain fatty acids, controlling intracellular lipid dynamics. FABP7 is expressed by astrocytes in the developing brain, and suggested to be involved in the control of astrocyte lipid homeostasis. In this study, we sought to examine the role of FABP7 in astrocytes, focusing on plasma membrane lipid raft function, which is important for receptor-mediated signal transduction in response to extracellular stimuli. In FABP7-knockout (KO) astrocytes, the ligand-dependent accumulation of Toll-like receptor 4 (TLR4) and glial cell-line-derived neurotrophic factor receptor alpha 1 into lipid raft was decreased, and the activation of mitogen-activated protein kinases and nuclear factor-κB was impaired after lipopolysaccharide (LPS) stimulation when compared with wild-type astrocytes. In addition, the expression of caveolin-1, not cavin-1, 2, 3, caveolin-2, and flotillin-1, was found to be decreased at the protein and transcriptional levels. FABP7 re-expression in FABP7-KO astrocytes rescued the decreased level of caveolin-1. Furthermore, caveolin-1-transfection into FABP7-KO astrocytes significantly increased TLR4 recruitment into lipid raft and tumor necrosis factor-α production after LPS stimulation. Taken together, these data suggest that FABP7 controls lipid raft function through the regulation of caveolin-1 expression and is involved in the response of astrocytes to the external stimuli. GLIA 2015;63:780-794.


Subject(s)
Astrocytes/cytology , Caveolae/metabolism , Caveolin 1/metabolism , Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation/genetics , Nerve Tissue Proteins/metabolism , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Caveolae/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Cholesterol/metabolism , Cytokines/metabolism , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , Signal Transduction/drug effects , Signal Transduction/genetics , Transduction, Genetic
7.
J Neural Transm (Vienna) ; 122(3): 477-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25005592

ABSTRACT

Disruption of synaptic networks has been advocated in the pathogenesis of psychiatric diseases like schizophrenia. The majority of synaptic proteins involved in neuronal communications are localized in lipid rafts. These rafts form the platform for coordinating neuronal signal transduction, by clustering interacting partners. The PAG1 protein is a transmembrane adaptor protein in the lipid raft signaling cluster that regulates Src family kinases (SFKs), a convergent point for multiple pathways regulating N-methyl-D-aspartate (NMDA) receptors. Reports of de novo missense mutations in PAG1 and SFK mediated reductions in tyrosine phosphorylation of NMDA receptor subunit proteins in schizophrenia patients, point to a putative role in schizophrenia pathogenesis. To evaluate this, we resequenced the entire coding region of PAG1 in Japanese schizophrenia patients (n = 1,140) and controls (n = 1,140). We identified eight missense variants, of which four were previously unreported. Case-control genetic association analysis of these variants in a larger cohort (n = 4,182) showed neither a statistically significant association of the individual variants with schizophrenia, nor any increased burden of the rare alleles in the patient group. Expression levels of PAG1 in post-mortem brain samples from schizophrenia patients and controls also showed no significant differences. To assess the precise role of PAG1 in schizophrenia, future studies with larger sample sizes are needed.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Brain/metabolism , Gene Expression Regulation/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation, Missense/genetics , Schizophrenia , Adult , Aged , Case-Control Studies , Cohort Studies , Exons/genetics , Female , Genetic Association Studies , Genetic Predisposition to Disease , Haplotypes , Humans , Japan , Male , Middle Aged , RNA, Messenger/metabolism , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/pathology
8.
Glycobiology ; 24(10): 926-34, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24821492

ABSTRACT

Myelin, a multilamellar structure extended from oligodendrocytes or Schwann cells, plays a critical role in maintenance of neuronal function, and damage or loss of myelin causes demyelinating diseases such as multiple sclerosis. For precise alignment of the myelin sheath, there is a requirement for expression of galactosylceramide (GalCer), a major glycosphingolipid in myelin. Synthesis of GalCer is strictly limited in oligodendrocytes in a developmental stage-specific manner. Ceramide galactosyltransferase (CGT), a key enzyme for biosynthesis of GalCer, exhibits restricted expression in oligodendrocytes but the mechanism is poorly understood. Based on our assumption that particular oligodendrocyte-lineage-specific transcription factors regulate CGT expression, we co-expressed a series of candidate transcription factors with the human CGT promoter-driving luciferase expression in oligodendroglioma cells to measure the promoter activity. We found that Nkx2.2 strongly activated the CGT promoter. In addition, we identified a novel repressive DNA element in the first intron of CGT and OLIG2, an oligodendrocyte-specific transcription factor, as a binding protein of this element. Moreover, overexpression of OLIG2 completely canceled the activating effect of Nkx2.2 on CGT promoter activity. Expression of CGT mRNA was also upregulated by Nkx2.2, but this upregulation was cancelled by co-expression of OLIG2 with Nkx2.2. Our study suggests that CGT expression is controlled by balanced expression of the negative modulator OLIG2 and positive regulator Nkx2.2, providing new insights into how expression of GalCer is tightly regulated in cell-type- and stage-specific manners.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Homeodomain Proteins/biosynthesis , Multiple Sclerosis/genetics , N-Acylsphingosine Galactosyltransferase/genetics , Nerve Tissue Proteins/biosynthesis , Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Galactosylceramides/biosynthesis , Galactosylceramides/metabolism , Gene Expression Regulation, Enzymologic/genetics , HeLa Cells , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Humans , Multiple Sclerosis/pathology , Myelin Sheath/metabolism , Myelin Sheath/pathology , N-Acylsphingosine Galactosyltransferase/biosynthesis , Nerve Tissue Proteins/genetics , Nuclear Proteins , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/enzymology , Oligodendroglia/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , Zebrafish Proteins
9.
J Pharmacol Sci ; 124(3): 294-300, 2014.
Article in English | MEDLINE | ID: mdl-24561447

ABSTRACT

Increasing evidence from the fields of neurophysiology and neuropathology has uncovered the role of polyunsaturated fatty acids (PUFA) in protecting neuronal cells from oxidative damage, controlling inflammation, regulating neurogenesis, and preserving neuronal function. Numerous epidemiological studies have shown that deficits in the dietary PUFA docosahexaenoic acid and eicosapentaenoic acid are associated with the onset and progression of neuropsychiatric illnesses such as dementia, schizophrenia, depression, and posttraumatic stress disorder (PTSD). Recent clinical trials have offered compelling evidence that suggests that n-3 PUFA could reduce depressive, psychotic, and suicidal symptoms, as well as aggression. Although many studies have had the validity of their results questioned because of small sample size, several studies have indicated that n-3 PUFA are useful therapeutic tools for the treatment of dementia, major depression, bipolar disorder, and PTSD. These findings suggest that the pharmacological and nutritional actions of n-3 PUFA may be beneficial in certain neuropsychiatric illnesses. This review article outlines the role of PUFA in neurodevelopment and the regulatory mechanisms in neuronal stem cell differentiation and also the possible use of PUFA as a prescription medicine for the prophylaxis or treatment of neuropsychiatric illnesses such as dementia, mood disorder, and PTSD.


Subject(s)
Fatty Acids, Omega-3/therapeutic use , Mental Disorders/drug therapy , Mental Disorders/prevention & control , Cell Differentiation/drug effects , Clinical Trials as Topic , Dementia/drug therapy , Dementia/prevention & control , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/physiology , Docosahexaenoic Acids/therapeutic use , Eicosapentaenoic Acid/pharmacology , Eicosapentaenoic Acid/physiology , Eicosapentaenoic Acid/therapeutic use , Fatty Acids, Omega-3/pharmacology , Fatty Acids, Omega-3/physiology , Humans , Mood Disorders/drug therapy , Mood Disorders/prevention & control , Neural Stem Cells/cytology , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/prevention & control
10.
Stem Cells ; 30(7): 1532-43, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22581784

ABSTRACT

New neurons are continually produced after birth from neural stem/progenitor cells (NSCs/NPCs) in the hippocampal dentate gyrus (DG). Recent studies have reported that fatty acid binding protein 7 (Fabp7/brain lipid binding protein (BLBP)) is required for the maintenance of embryonic NSCs/NPCs and have identified an association between the Fabp7 gene and behavioral paradigms that correlate with hippocampal functions. However, the specific roles of Fabps in postnatal neurogenesis remain unknown. Herein, we demonstrate the effects of Fabp7, and another Fabp, Fabp5, on postnatal neurogenesis. Fabp7 and Fabp5 were detected in the subgranular zone (SGZ) of the DG, and Fabp7+ cells were less differentiated than Fabp5+ cells. We analyzed the differentiation state of NSCs/NPCs in the SGZ of 4-week-old (4w) Fabp7 knockout (7KO), Fabp5 KO (5KO), and Fabp7/Fabp5 double KO (7/5KO) mice and found that the number of NSCs/NPCs was dramatically reduced compared with wild-type mice. Although the uptake of BrdU 1 day after injection was decreased in all KO mice, the survival of BrdU+ cells 1 month after injection was increased in the 7/5KO mice compared to other three genotypes. We also observed an enhancement of neuronal differentiation in all Fabp KO mice. In addition, the proliferation and survival of NSCs/NPCs differed along the anterior-posterior axis (A-P axis). A greater number of newborn cells in the posterior region became extinct, but this tendency was not apparent in the Fabps KO mice. These data suggest that Fabp7 and Fabp5 have differential roles for proliferation and survival of the NSCs/NPCs during postnatal DG neurogenesis.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Fatty Acid-Binding Proteins/poisoning , Hippocampus/cytology , Neoplasm Proteins/poisoning , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins/genetics , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Neurogenesis/genetics
11.
Nihon Yakurigaku Zasshi ; 158(3): 238-241, 2023 May 01.
Article in Japanese | MEDLINE | ID: mdl-36990793

ABSTRACT

Our previous study has suggested that peroxisome proliferator-activated receptor α (PPARα) plays a crucial role in the pathophysiology of schizophrenia. In the current study, we screened and identified rare variants in the PPARA gene (encoding PPARα) of schizophrenia subjects. In vitro study showed that those variants decreased activities of PPARα as a transcription factor. Ppara KO mice exhibited a deficit in the sensorimotor gating function and schizophrenia-related histological abnormalities. RNA-seq analysis revealed that PPARα regulates the expression of synaptogenesis signaling pathway-related genes in the brain. Remarkably, treatment of mice with the PPARα agonist fenofibrate alleviated an NMDA receptor antagonist, phencyclidine (PCP)-induced spine pathology and reduced sensitivity to MK-801, another NMDA receptor antagonist. In conclusion, the current study further supports the idea that perturbation in the PPARα-regulated transcriptional machinery leads to a predisposition to schizophrenia, probably by affecting synapse physiology. This study also demonstrates that PPARα can serve as a novel therapeutic target for schizophrenia.


Subject(s)
Fenofibrate , PPAR alpha , Schizophrenia , Animals , Mice , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , PPAR alpha/genetics , PPAR alpha/agonists , PPAR alpha/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, N-Methyl-D-Aspartate , Schizophrenia/drug therapy , Schizophrenia/genetics
12.
FEBS J ; 290(7): 1798-1821, 2023 04.
Article in English | MEDLINE | ID: mdl-36325660

ABSTRACT

Fatty acid-binding protein 7 (FABP7), one of the fatty acid (FA) chaperones involved in the modulation of intracellular FA metabolism, is highly expressed in glioblastoma, and its expression is associated with decreased patients' prognosis. Previously, we demonstrated that FABP7 requires its binding partner to exert its function and that a mutation in the FA-binding site of FABP7 affects tumour biology. Here, we explored the role of FA ligand binding for FABP7 function in tumour proliferation and examined the mechanism of FABP7 and ligand interaction in tumour biology. We discovered that among several FA treatment, oleic acid (OA) boosted cell proliferation of FABP7-expressing cells. In turn, OA increased FABP7 nuclear localization, and the accumulation of FABP7-OA complex in the nucleus induced the formation of nuclear lipid droplet (nLD), as well as an increase in colocalization of nLD with promyelocytic leukaemia (PML) nuclear bodies. Furthermore, OA increased mRNA levels of proliferation-related genes in FABP7-expressing cells through histone acetylation. Interestingly, these OA-boosted functions were abrogated in FABP7-knockout cells and mutant FABP7-overexpressing cells. Thus, our findings suggest that FABP7-OA intracellular interaction may modulate nLD formation and the epigenetic status thereby enhancing transcription of proliferation-regulating genes, ultimately driving tumour cell proliferation.


Subject(s)
Glioma , Oleic Acid , Humans , Fatty Acid-Binding Protein 7/genetics , Fatty Acid-Binding Protein 7/metabolism , Oleic Acid/pharmacology , Oleic Acid/metabolism , Lipid Droplets/metabolism , Ligands , Glioma/pathology , Cell Proliferation , Tumor Suppressor Proteins/genetics
14.
Hum Genet ; 131(3): 443-51, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21927946

ABSTRACT

We recently reported the results of a genome-wide association study (GWAS) of schizophrenia in the Japanese population. In that study, a single nucleotide polymorphism (SNP) (rs3106653) in the KCNJ3 (potassium inwardly rectifying channel, subfamily J, member 3) gene located at 2q24.1 showed association with schizophrenia in two independent sample sets. KCNJ3, also termed GIRK1 or Kir3.1, is a member of the G protein-activated inwardly rectifying K(+) channel (GIRK) group. GIRKs are widely distributed in the brain and play an important role in regulating neural excitability through the activation of various G protein-coupled receptors. In this study, we set out to examine this association using a different population. We first performed a gene-centric association study of the KCNJ3 gene, by genotyping 38 tagSNPs in the Chinese population. We detected nine SNPs that displayed significant association with schizophrenia (lowest P = 0.0016 for rs3106658, Global significance = 0.036). The initial marker SNP (rs3106653) examined in our prior GWAS in the Japanese population also showed nominally significant association in the Chinese population (P = 0.028). Next, we analyzed transcript levels in the dorsolateral prefrontal cortex of postmortem brains from patients with schizophrenia and bipolar disorder and from healthy controls, using real-time quantitative RT-PCR. We found significantly lower KCNJ3 expression in postmortem brains from schizophrenic and bipolar patients compared with controls. These data suggest that the KCNJ3 gene is genetically associated with schizophrenia in Asian populations and add further evidence to the "channelopathy theory of psychiatric illnesses".


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Schizophrenia/genetics , Adult , Asian People/genetics , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide
15.
Genes Cells ; 16(7): 778-90, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21668588

ABSTRACT

Arachidonic acid (ARA) and docosahexaenoic acid (DHA), which are the dominant polyunsaturated fatty acids in the brain, have crucial roles in brain development and function. Recent studies have shown that ARA and DHA promote postnatal neurogenesis. However, the direct effects of ARA on neural stem/progenitor cells (NSPCs) and the effects of ARA and DHA on NSPCs at the neurogenic and subsequent gliogenic stages are still unknown. Here, we analyzed the effects of ARA and DHA on neurogenesis, specifically maintenance and differentiation, using neurosphere assays. We confirmed that primary neurospheres are neurogenic NSPCs and that tertiary neurospheres are gliogenic NSPCs. Regarding the effects of ARA and DHA on neurogenic NSPCs, ARA and DHA increased the number of neurospheres, whereas neither ARA nor DHA had a detectable effect on NSPCs in the differentiation condition. In gliogenic NSPCs, DHA increased the number of neurospheres, whereas ARA had no such effect. In contrast, ARA increased the number of astrocytes, whereas DHA increased the number of neurons in the differentiation condition. These results suggest that ARA promotes the maintenance of neurogenic NSPCs and might induce the glial differentiation of gliogenic NSPCs and that DHA promotes the maintenance of both neurogenic and gliogenic NSPCs and might lead to the neuronal differentiation of gliogenic NSPCs.


Subject(s)
Arachidonic Acid/pharmacology , Docosahexaenoic Acids/pharmacology , Neural Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Neural Stem Cells/cytology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
16.
Histochem Cell Biol ; 136(5): 501-13, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21938553

ABSTRACT

Reactive gliosis, in which astrocytes as well as other types of glial cells undergo massive proliferation, is a common hallmark of all brain pathologies. Brain-type fatty acid-binding protein (FABP7) is abundantly expressed in neural stem cells and astrocytes of developing brain, suggesting its role in differentiation and/or proliferation of glial cells through regulation of lipid metabolism and/or signaling. However, the role of FABP7 in proliferation of glial cells during reactive gliosis is unknown. In this study, we examined the expression of FABP7 in mouse cortical stab injury model and also the phenotype of FABP7-KO mice in glial cell proliferation. Western blotting showed that FABP7 expression was increased significantly in the injured cortex compared with the contralateral side. By immunohistochemistry, FABP7 was localized to GFAP(+) astrocytes (21% of FABP7(+) cells) and NG2(+) oligodendrocyte progenitor cells (62%) in the normal cortex. In the injured cortex there was no change in the population of FABP7(+)/NG2(+) cells, while there was a significant increase in FABP7(+)/GFAP(+) cells. In the stab-injured cortex of FABP7-KO mice there was decrease in the total number of reactive astrocytes and in the number of BrdU(+) astrocytes compared with wild-type mice. Primary cultured astrocytes from FABP7-KO mice also showed a significant decrease in proliferation and omega-3 fatty acid incorporation compared with wild-type astrocytes. Overall, these data suggest that FABP7 is involved in the proliferation of astrocytes by controlling cellular fatty acid homeostasis.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/injuries , Cerebral Cortex/metabolism , Fatty Acid-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Wounds, Stab/metabolism , Animals , Animals, Newborn , Antigens/metabolism , Astrocytes/pathology , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins/genetics , Fatty Acids, Omega-3/metabolism , Gene Expression , Gliosis/metabolism , Gliosis/pathology , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , Proteoglycans/metabolism , Wounds, Stab/genetics , Wounds, Stab/pathology
17.
Am J Med Genet B Neuropsychiatr Genet ; 156B(5): 581-92, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21563301

ABSTRACT

Many studies have suggested that myelin dysfunction may be causally involved in the pathogenesis of schizophrenia. Nogo (RTN4), myelin-associated glycoprotein (MAG) and oligodendrocyte myelin glycoprotein (OMG) all bind to the common receptor, Nogo-66 receptor 1 (RTN4R). We examined 68 single nucleotide polymorphisms (SNPs) (51 with genotyping and 17 with imputation analysis) from these four genes for genetic association with schizophrenia, using a 2,120 case-control sample from the Japanese population. Allelic tests showed nominally significant association of two RTN4 SNPs (P = 0.047 and 0.037 for rs11894868 and rs2968804, respectively) and two MAG SNPs (P = 0.034 and 0.029 for rs7249617 and rs16970218, respectively) with schizophrenia. The MAG SNP rs7249617 also showed nominal significance in a genotypic test (P = 0.017). In haplotype analysis, the MAG haplotype block including rs7249617 and rs16970218 showed nominal significance (P = 0.008). These associations did not remain significant after correction for multiple testing, possibly due to their small genetic effect. In the imputation analysis of RTN4, the untyped SNP rs2972090 showed nominally significant association (P = 0.032) and several imputed SNPs showed marginal associations. Moreover, in silico analysis (PolyPhen) of a missense variant (rs11677099: Asp357Val), which is in strong linkage disequilibrium with rs11894868, predicted a deleterious effect on Nogo protein function. Despite a failure to detect robust associations in this Japanese cohort, our nominally positive signals, taken together with previously reported biological and genetic findings, add further support to the "disturbed myelin system theory of schizophrenia" across different populations.


Subject(s)
Myelin Proteins/genetics , Myelin-Associated Glycoprotein/genetics , Receptors, Cell Surface/genetics , Schizophrenia/genetics , Adult , Aged , Case-Control Studies , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Haplotypes , Humans , Japan , Male , Middle Aged , Myelin Proteins/metabolism , Myelin-Associated Glycoprotein/metabolism , Nogo Proteins , Nogo Receptor 1 , Polymorphism, Single Nucleotide , Receptors, Cell Surface/metabolism
18.
Schizophr Bull ; 47(4): 1190-1200, 2021 07 08.
Article in English | MEDLINE | ID: mdl-33595068

ABSTRACT

We previously identified quantitative trait loci (QTL) for prepulse inhibition (PPI), an endophenotype of schizophrenia, on mouse chromosome 10 and reported Fabp7 as a candidate gene from an analysis of F2 mice from inbred strains with high (C57BL/6N; B6) and low (C3H/HeN; C3H) PPI levels. Here, we reanalyzed the previously reported QTLs with increased marker density. The highest logarithm of odds score (26.66) peaked at a synonymous coding and splice-site variant, c.753G>A (rs257098870), in the Cdh23 gene on chromosome 10; the c.753G (C3H) allele showed a PPI-lowering effect. Bayesian multiple QTL mapping also supported the same variant with a posterior probability of 1. Thus, we engineered the c.753G (C3H) allele into the B6 genetic background, which led to dampened PPI. We also revealed an e-QTL (expression QTL) effect imparted by the c.753G>A variant for the Cdh23 expression in the brain. In a human study, a homologous variant (c.753G>A; rs769896655) in CDH23 showed a nominally significant enrichment in individuals with schizophrenia. We also identified multiple potentially deleterious CDH23 variants in individuals with schizophrenia. Collectively, the present study reveals a PPI-regulating Cdh23 variant and a possible contribution of CDH23 to schizophrenia susceptibility.


Subject(s)
Cadherin Related Proteins/genetics , Cadherins/genetics , Prepulse Inhibition/genetics , Schizophrenia/genetics , Alleles , Animals , Humans , Mice , Quantitative Trait Loci
19.
EMBO Mol Med ; 13(4): e12574, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33656268

ABSTRACT

Genomic defects with large effect size can help elucidate unknown pathologic architecture of mental disorders. We previously reported on a patient with schizophrenia and a balanced translocation between chromosomes 4 and 13 and found that the breakpoint within chromosome 4 is located near the LDB2 gene. We show here that Ldb2 knockout (KO) mice displayed multiple deficits relevant to mental disorders. In particular, Ldb2 KO mice exhibited deficits in the fear-conditioning paradigm. Analysis of the amygdala suggested that dysregulation of synaptic activities controlled by the immediate early gene Arc is involved in the phenotypes. We show that LDB2 forms protein complexes with known transcription factors. Consistently, ChIP-seq analyses indicated that LDB2 binds to > 10,000 genomic sites in human neurospheres. We found that many of those sites, including the promoter region of ARC, are occupied by EGR transcription factors. Our previous study showed an association of the EGR family genes with schizophrenia. Collectively, the findings suggest that dysregulation in the gene expression controlled by the LDB2-EGR axis underlies a pathogenesis of subset of mental disorders.


Subject(s)
Schizophrenia , Animals , Fear , Gene Expression , Humans , LIM Domain Proteins/genetics , Mice , Mice, Knockout , Schizophrenia/genetics , Transcription Factors/genetics
20.
J Neurochem ; 115(6): 1374-85, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20977478

ABSTRACT

Deficits in prepulse inhibition (PPI) are known in mental illnesses, including schizophrenia. NMDA receptor function affects PPI integrity and D-serine and glycine are endogenous co-agonists for the receptor. Our previous quantitative trait loci analysis using C57BL/6 (B6) mice with better PPI performance and C3H/He (C3) with lower PPI score, shows that genes for both D-serine synthesizing enzyme and enzyme for reversible conversion between glycine and L-serine (Srr and Shmt1, respectively) are located in the same PPI-quantitative trait loci peak. Therefore, we set out to determine which gene is likely to explain the PPI difference and whether the gene is potentially relevant to schizophrenia. We first examined brain interstitial fluid levels of the two amino acids using microdialysis. Recovery of D-serine and glycine from the dialysate was higher in B6, compared to C3. Next, we analyzed expression levels and genetic polymorphisms of the two genes. There were promoter polymorphisms in Shmt1, which elicit lower transcriptional activity in B6 compared to C3 conforming to the results of brain expression levels, but no functional genetic variants in Srr. Finally, we evaluated expression levels of the two genes in the postmortem brains of schizophrenia and genetic associations with the disease. The SHMT1 levels were higher in schizophrenic brains compared to controls, but no changes in SRR levels. We detected a nominal association between SHMT1 and schizophrenia. These results suggest that Shmt1 (SHMT1), but not Srr, is likely to be one of the genetic components regulating PPI in mice and possibly relevant to schizophrenia.


Subject(s)
Disease Models, Animal , Glycine Hydroxymethyltransferase/genetics , Neural Inhibition/genetics , Reflex, Startle/physiology , Schizophrenia/genetics , Schizophrenia/physiopathology , Adult , Animals , Base Sequence , Female , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Middle Aged , Molecular Sequence Data , Racemases and Epimerases/genetics , Reflex, Startle/genetics , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL