Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Kunio Miyake is active.

Publication


Featured researches published by Kunio Miyake.


Clinical Epigenetics | 2012

Epigenetic understanding of gene-environment interactions in psychiatric disorders: a new concept of clinical genetics

Takeo Kubota; Kunio Miyake; Takae Hirasawa

Epigenetics is a mechanism that regulates gene expression independently of the underlying DNA sequence, relying instead on the chemical modification of DNA and histone proteins. Although environmental and genetic factors were thought to be independently associated with disorders, several recent lines of evidence suggest that epigenetics bridges these two factors. Epigenetic gene regulation is essential for normal development, thus defects in epigenetics cause various rare congenital diseases. Because epigenetics is a reversible system that can be affected by various environmental factors, such as drugs, nutrition, and mental stress, the epigenetic disorders also include common diseases induced by environmental factors. In this review, we discuss the nature of epigenetic disorders, particularly psychiatric disorders, on the basis of recent findings: 1) susceptibility of the conditions to environmental factors, 2) treatment by taking advantage of their reversible nature, and 3) transgenerational inheritance of epigenetic changes, that is, acquired adaptive epigenetic changes that are passed on to offspring. These recently discovered aspects of epigenetics provide a new concept of clinical genetics.


BMC Neuroscience | 2011

The protocadherins, PCDHB1 and PCDH7, are regulated by MeCP2 in neuronal cells and brain tissues: implication for pathogenesis of Rett syndrome

Kunio Miyake; Takae Hirasawa; Masaki Soutome; Masayuki Itoh; Yu-ichi Goto; Kazushi Endoh; Kenichiro Takahashi; Shinichi Kudo; Takayuki Nakagawa; Sana Yokoi; Takahiro Taira; Johji Inazawa; Takeo Kubota

BackgroundRett syndrome is a neurodevelopmental and autistic disease caused by mutations of Methyl-CpG-binding protein 2 (MECP2) gene. MeCP2 protein is mainly expressed in neurons and binds to methylated gene promoters to suppress their expression, indicating that Rett syndrome is caused by the deregulation of target genes in neurons. However, it is likely that there are more unidentified neuronal MeCP2-targets associated with the neurological features of RTT.ResultsUsing a genome-microarray approach, we found 22 genomic regions that contain sites potentially regulated by MeCP2 based on the features of MeCP2 binding, DNA methylation, and repressive histone modification in human cell lines. Within these regions, Chromatin immunoprecipitation (ChIP) analysis revealed that MeCP2 binds to the upstream regions of the protocadherin genes PCDHB1 and PCDH7 in human neuroblastoma SH-SY5Y cells. PCDHB1 and PCDH7 promoter activities were down-regulated by MeCP2, but not by MBD-deleted MeCP2. These gene expression were up-regulated following MeCP2 reduction with siRNA in SH-SY5Y cells and in the brains of Mecp2-null mice. Furthermore, PCDHB1 was up-regulated in postmortem brains from Rett syndrome patients.ConclusionsWe identified MeCP2 target genes that encode neuronal adhesion molecules using ChIP-on-BAC array approach. Since these protocadherin genes are generally essential for brain development, aberrant regulation of these molecules may contribute to the pathogenesis of the neurological features observed in Rett syndrome.


Advances in Experimental Medicine and Biology | 2012

Epigenetics in Autism and Other Neurodevelopmental Diseases

Kunio Miyake; Takae Hirasawa; Tsuyoshi Koide; Takeo Kubota

Autism was previously thought to be caused by environmental factors. However, genetic factors are now considered to be more contributory to the pathogenesis of autism, based on the recent findings of mutations in the genes which encode synaptic molecules associated with the communication between neurons. Epigenetic is a mechanism that controls gene expression without changing DNA sequence but by changing chromosomal histone modifications and its abnormality is associated with several neurodevelopmental diseases. Since epigenetic modifications are known to be affected by environmental factors such as nutrition, drugs and mental stress, autistic diseases are not only caused by congenital genetic defects, but may also be caused by environmental factors via epigenetic mechanism. In this chapter, we introduce autistic diseases caused by epigenetic failures and discuss epigenetic changes by environmental factors and discuss new treatments for neurodevelopmental diseases based on the recent epigenetic findings.


Molecular Brain | 2015

Differentiation of multipotent neural stem cells derived from Rett syndrome patients is biased toward the astrocytic lineage

Tomoko Andoh-Noda; Wado Akamatsu; Kunio Miyake; Takuya Matsumoto; Ryo Yamaguchi; Tsukasa Sanosaka; Yohei Okada; Tetsuro Kobayashi; Manabu Ohyama; Kinichi Nakashima; Hiroshi Kurosawa; Takeo Kubota; Hideyuki Okano

BackgroundRett syndrome (RTT) is one of the most prevalent neurodevelopmental disorders in females, caused by de novo mutations in the X-linked methyl CpG-binding protein 2 gene, MECP2. Although abnormal regulation of neuronal genes due to mutant MeCP2 is thought to induce autistic behavior and impaired development in RTT patients, precise cellular mechanisms underlying the aberrant neural progression remain unclear.ResultsTwo sets of isogenic pairs of either wild-type or mutant MECP2-expressing human induced pluripotent stem cell (hiPSC) lines were generated from a single pair of 10-year-old RTT-monozygotic (MZ) female twins. Mutant MeCP2-expressing hiPSC lines did not express detectable MeCP2 protein during any stage of differentiation. The lack of MeCP2 reflected altered gene expression patterns in differentiated neural cells rather than in undifferentiated hiPSCs, as assessed by microarray analysis. Furthermore, MeCP2 deficiency in the neural cell lineage increased astrocyte-specific differentiation from multipotent neural stem cells. Additionally, chromatin immunoprecipitation (ChIP) and bisulfite sequencing assays indicated that anomalous glial fibrillary acidic protein gene (GFAP) expression in the MeCP2-negative, differentiated neural cells resulted from the absence of MeCP2 binding to the GFAP gene.ConclusionsAn isogenic RTT-hiPSC model demonstrated that MeCP2 participates in the differentiation of neural cells. Moreover, MeCP2 deficiency triggers perturbation of astrocytic gene expression, yielding accelerated astrocyte formation from RTT-hiPSC-derived neural stem cells. These findings are likely to shed new light on astrocytic abnormalities in RTT, and suggest that astrocytes, which are required for neuronal homeostasis and function, might be a new target of RTT therapy.


Neurochemistry International | 2007

Phosphorylation of methyl-CpG binding protein 2 (MeCP2) regulates the intracellular localization during neuronal cell differentiation

Kunio Miyake; Kaoru Nagai

Methyl-CpG binding protein 2 (MeCP2) is a transcriptional repressor which recognizes methylated CpG dinucleotides. Mutations in the MeCP2 gene is known to cause human autistic disease Rett syndrome, but its molecular mechanisms remain to be elucidated. Since MeCP2 is a DNA-binding protein, it has been believed that MeCP2 functions only in the nucleus. We herein show that MeCP2 is localized in the cytosol as well as in the nucleus of neuronal cells. Through the use of immunofluorescence and Western blot analyses, MeCP2 was found to be localized both in the nucleus and cytosol of rat PC-12 and mouse Neuro2a cells before neuronal differentiation, and it was translocated into the nucleus during differentiation. In primary cultured neurons from mouse cortex, MeCP2 was expressed in whole cell bodies on the first day of culture while after 7 days of culture, MeCP2 was localized mainly in the nucleus. Furthermore, MeCP2 was re-localized in the nucleus and cytosol after 14 days of culture. To study the molecular mechanisms of translocation, we analyzed the post-translational modification of MeCP2. The cytosolic MeCP2 was Ser/Thr-phosphorylated, while the nuclear MeCP2 was not. Both the cytosolic and nuclear MeCP2 were SUMOylated, which has been reported to be a nuclear transport signal. Our data suggests that the nuclear translocation of neuronal MeCP2 was induced during differentiation and/or maturation, and that Ser/Thr-phosphorylation regulates its translocation.


PLOS ONE | 2014

Expressions of Tight Junction Proteins Occludin and Claudin-1 Are under the Circadian Control in the Mouse Large Intestine: Implications in Intestinal Permeability and Susceptibility to Colitis

Kyoko Oh-oka; Hiroshi Kono; Kayoko Ishimaru; Kunio Miyake; Takeo Kubota; Hideoki Ogawa; Ko Okumura; Shigenobu Shibata; Atsuhito Nakao

Background & Aims The circadian clock drives daily rhythms in behavior and physiology. A recent study suggests that intestinal permeability is also under control of the circadian clock. However, the precise mechanisms remain largely unknown. Because intestinal permeability depends on tight junction (TJ) that regulates the epithelial paracellular pathway, this study investigated whether the circadian clock regulates the expression levels of TJ proteins in the intestine. Methods The expression levels of TJ proteins in the large intestinal epithelium and colonic permeability were analyzed every 4, 6, or 12 hours between wild-type mice and mice with a mutation of a key clock gene Period2 (Per2; mPer2m/m). In addition, the susceptibility to dextran sodium sulfate (DSS)-induced colitis was compared between wild-type mice and mPer2m/m mice. Results The mRNA and protein expression levels of Occludin and Claudin-1 exhibited daily variations in the colonic epithelium in wild-type mice, whereas they were constitutively high in mPer2m/m mice. Colonic permeability in wild-type mice exhibited daily variations, which was inversely associated with the expression levels of Occludin and Claudin-1 proteins, whereas it was constitutively low in mPer2m/m mice. mPer2m/m mice were more resistant to the colonic injury induced by DSS than wild-type mice. Conclusions Occludin and Claudin-1 expressions in the large intestine are under the circadian control, which is associated with temporal regulation of colonic permeability and also susceptibility to colitis.


Journal of Clinical Immunology | 2015

A Female Patient with Incomplete Hemophagocytic Lymphohistiocytosis Caused by a Heterozygous XIAP Mutation Associated with Non-Random X-Chromosome Inactivation Skewed Towards the Wild-Type XIAP Allele

Xi Yang; Akihiro Hoshino; Takashi Taga; Tomoaki Kunitsu; Yuhachi Ikeda; Takahiro Yasumi; Kenichi Yoshida; Taizo Wada; Kunio Miyake; Takeo Kubota; Yusuke Okuno; Hideki Muramatsu; Yuichi Adachi; Satoru Miyano; Seishi Ogawa; Seiji Kojima; Hirokazu Kanegane

X-linked lymphoproliferative disease (XLP) is a rare inherited immunodeficiency that often leads to hemophagocytic lymphohistiocytosis (HLH). XLP can be classified as XLP1 or XLP2, caused by mutations in SH2D1A and XIAP, respectively. In women, X-chromosome inactivation (XCI) of most X-linked genes occurs on one of the X chromosomes in each cell. The choice of which X chromosome remains activated is generally random, although genetic differences and selective advantage may cause one of the X chromosomes to be preferentially inactivated. Here we describe three patients with pancytopenia, including one female patient, in a Japanese family with a novel XIAP mutation. All three patients exhibited deficient XIAP protein expression, impaired NOD2/XIAP signaling, and augmented activation-induced cell death. In the female patient, the paternally derived X chromosome was non-randomly and exclusively inactivated in her peripheral blood and hair root cells. In contrast to asymptomatic females, this patient exhibied non-random XCI skewed towards the wild-type XIAP allele. This is the first report of a female patient with incomplete HLH resulting from a heterozygous XIAP mutation in association with non-random XCI.


Comparative and Functional Genomics | 2017

Neurodevelopmental Disorders and Environmental Toxicants: Epigenetics as an Underlying Mechanism

Nguyen Quoc Vuong Tran; Kunio Miyake

The increasing prevalence of neurodevelopmental disorders, especially autism spectrum disorders (ASD) and attention deficit hyperactivity disorder (ADHD), calls for more research into the identification of etiologic and risk factors. The Developmental Origin of Health and Disease (DOHaD) hypothesizes that the environment during fetal and childhood development affects the risk for many chronic diseases in later stages of life, including neurodevelopmental disorders. Epigenetics, a term describing mechanisms that cause changes in the chromosome state without affecting DNA sequences, is suggested to be the underlying mechanism, according to the DOHaD hypothesis. Moreover, many neurodevelopmental disorders are also related to epigenetic abnormalities. Experimental and epidemiological studies suggest that exposure to prenatal environmental toxicants is associated with neurodevelopmental disorders. In addition, there is also evidence that environmental toxicants can result in epigenetic alterations, notably DNA methylation. In this review, we first focus on the relationship between neurodevelopmental disorders and environmental toxicants, in particular maternal smoking, plastic-derived chemicals (bisphenol A and phthalates), persistent organic pollutants, and heavy metals. We then review studies showing the epigenetic effects of those environmental factors in humans that may affect normal neurodevelopment.


Journal of Biological Chemistry | 2013

Oligodendrocyte Transcription Factor 1 (Olig1) Is a Smad Cofactor Involved in Cell Motility Induced by Transforming Growth Factor-β

Mitsuyoshi Motizuki; Kazunobu Isogaya; Kunio Miyake; Hiroaki Ikushima; Takeo Kubota; Kohei Miyazono; Masao Saitoh; Keiji Miyazawa

Background: To date, the Smad cofactor involved in cell motility induced by transforming growth factor-β (TGF-β) has not been identified. Results: Knockdown of oligodendrocyte transcription factor-1 (Olig1), as well as inhibition of the Olig1-Smad interaction, resulted in attenuation of TGF-β-induced cell motility. Conclusion: Olig1 is involved in TGF-β-induced cell motility. Significance: This study enhances understanding of the regulation of TGF-β-induced cell motility. Transforming growth factor (TGF)-β plays crucial roles in embryonic development and adult tissue homeostasis by eliciting various cellular responses in target cells. TGF-β signaling is principally mediated through receptor-activated Smad proteins, which regulate expression of target genes in cooperation with other DNA-binding transcription factors (Smad cofactors). In this study, we found that the basic helix-loop-helix transcription factor Olig1 is a Smad cofactor involved in TGF-β-induced cell motility. Knockdown of Olig1 attenuated TGF-β-induced cell motility in chamber migration and wound healing assays. In contrast, Olig1 knockdown had no effect on bone morphogenetic protein-induced cell motility, TGF-β-induced cytostasis, or epithelial-mesenchymal transition. Furthermore, we observed that cooperation of Smad2/3 with Olig1 is regulated by a peptidyl-prolyl cis/trans-isomerase, Pin1. TGF-β-induced cell motility, induction of Olig1-regulated genes, and physical interaction between Smad2/3 and Olig1 were all inhibited after knockdown of Pin1, indicating a novel mode of regulation of Smad signaling. We also found that Olig1 interacts with the L3 loop of Smad3. Using a synthetic peptide corresponding to the L3 loop of Smad3, we succeeded in selectively inhibiting TGF-β-induced cell motility. These findings may lead to a new strategy for selective regulation of TGF-β-induced cellular responses.


Epigenomics | 2013

Role of epigenetics in Rett syndrome

Takeo Kubota; Kunio Miyake; Takae Hirasawa

Rett syndrome (RTT) is an X-linked neurodevelopmental disease caused by MECP2 mutations. The MeCP2 protein was originally thought to function as a transcription repressor by binding to methylated CpG dinucleotides, but is now also thought to be a transcription activator. Recent studies suggest that MeCP2 is not only being expressed in neurons, but also in glial cells, which suggests a new paradigm for understanding the pathogenesis of RTT. It has also been demonstrated that reintroduction of MeCP2 into behaviorally affected Mecp2-null mice after birth rescues neurological symptoms, which indicates that epigenetic failures in RTT are reversible. Therefore, RTT may well be seen as a model disease that can be potentially treated by taking advantage of the reversibility of epigenetic phenomena in various congenital neurodevelopmental diseases that were previously thought to be untreatable.

Collaboration


Dive into the Kunio Miyake's collaboration.

Top Co-Authors

Avatar

Takeo Kubota

University of Yamanashi

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kaoru Nagai

University of Yamanashi

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kanji Sugita

University of Yamanashi

View shared research outputs
Top Co-Authors

Avatar

Kumiko Goi

University of Yamanashi

View shared research outputs
Researchain Logo
Decentralizing Knowledge