宮田 信吾(ミヤタ シンゴ)

東洋医学研究所教授

Last Updated :2024/06/18

■教員コメント

コメント

脳の病気、こころの病気の研究および漢方の臨床応用研究を行っています。特に、神経変性疾患やストレス性精神疾患発症の分子機序の研究と、新規治療薬や診断薬開発のための植物成分の検討です。

■研究者基本情報

学位

  • 博士(医学)(大阪大学)

科研費研究者番号

70403194

プロフィール

  • うつ病などの精神神経症状の発症機構、脳におけるタンパクメチル化の意義、オリゴデンドロサイトの機能解析、精神神経症状への漢方薬の作用機序、Neuron-Glia相互作用と脳機能、ホヤ神経系を用いた研究、小胞体ストレス・ゴルジストレスの分子機序などの研究を行い、それぞれの科学的基盤形成を目指しています。Thank you for your everything... I am full of gratitude!!!

研究キーワード

  • 細胞接着分子   創薬   microRNA   視床下部   拡散テンソル   髄鞘   不安障害   統合失調症   漢方薬   局所的翻訳   樹状突起   包括脳ネットワーク   大脳皮質   海馬   神経回路   ゴルジ体   ストレス   オリゴデンドロサイト   うつ病   CARM1   マイクログリア   脊髄損傷   PRMT1   protein methylation   PRMT8   

現在の研究分野(キーワード)

脳の病気、こころの病気の研究および漢方の臨床応用研究を行っています。特に、神経変性疾患やストレス性精神疾患発症の分子機序の研究と、新規治療薬や診断薬開発のための植物成分の検討です。

研究分野

  • ライフサイエンス / 発生生物学
  • ライフサイエンス / 分子生物学
  • ライフサイエンス / 細胞生物学
  • ライフサイエンス / 神経形態学
  • ライフサイエンス / 解剖学
  • ライフサイエンス / 神経科学一般

■経歴

経歴

  • 2016年04月 - 現在  近畿大学東洋医学研究所 分子脳科学研究部門(基礎研究部門)教授
  • 2012年04月 - 2016年03月  近畿大学東洋医学研究所 分子脳科学研究部門准教授
  • 2006年04月 - 2012年03月  大阪大学 子どものこころの分子統御機構研究センター 特任助教(兼任)
  • 2005年04月 - 2012年03月  大阪大学医学系研究科 神経機能形態学講座(旧 第二解剖)助教
  • 2002年04月 - 2005年03月  日本学術振興会特別研究員(DC1)

学歴

  • 2001年04月 - 2005年03月   大阪大学   大学院医学系研究科   博士課程

委員歴

  • 2023年05月 - 現在   Frontiers in Psychiatry   Guest Associate Editor
  • 2023年03月 - 現在   Neurology International   Editorial Board
  • 2023年02月 - 現在   日本ミエリン研究会   世話人
  • 2022年 - 現在   Frontiers in Cellular Neuroscience, Non-Neuronal Cells section   Permanent Associate Editor
  • 2017年 - 現在   日本生化学会 近畿支部   幹事
  • 2017年 - 現在   日本解剖学会   代議員
  • 2017年 - 現在   Osaka Science-based Kampo 研究会   世話人
  • 2016年 - 現在   日本生化学会   評議員
  • 2016年 - 現在   日本自律神経学会   評議員
  • 2013年 - 現在   日本神経化学会   評議員
  • 2023年11月 - 2025年03月   第130回日本解剖学会・第102回日本生理学会・第98回日本薬理学会 合同大会(APPW2025)   プログラム委員
  • 2023年05月 - 2024年03月   日本神経化学会   研究助成金等候補者選考委員
  • 2022年10月 - 2024年03月   Frontiers in Cellular Neuroscience   Research Topic Editor
  • 2022年09月 - 2023年08月   日本生化学会近畿支部   近畿支部奨励賞審査委員

■研究活動情報

受賞

  • 2016年 近畿大学 医学会賞
     
    受賞者: 宮田信吾
  • 2014年 近畿大学 医学会賞
     
    受賞者: 宮田信吾
  • 2013年 近畿大学医学会 一般演題優秀賞
     
    受賞者: 宮田信吾
  • 2000年 日本薬学会 第120年会 講演ハイライト 採択
     
    受賞者: 宮田信吾

論文

  • Editorial: Oligodendrocytes: from their development to function and dysfunction.
    Miyata S; Wake H
    Frontiers in Cellular Neuroscience 18 1376931  2024年03月 [査読有り][招待有り]
  • 急性病態における意識障害・認知機能障害と漢方薬
    宮田 信吾
    ICUとCCU 48 3 137 - 143 2024年03月 [査読有り][招待有り]
  • 脳の発生および構造から見たアルギニン残基のメチル化制御
    宮田 信吾
    生化学 96 1 86 - 90 2024年03月 [査読有り][招待有り]
  • 清水 尚子; 石野 雄吾; 武田 卓; 遠山 正彌; 宮田 信吾
    産婦人科の実際 72 3 231 - 235 金原出版(株) 2023年03月 
    <文献概要>女性の更年期障害は,卵巣機能低下によるエストロゲン分泌減少などの生物学的要因に加え,社会心理的ストレスなどの環境要因が複雑に関与し,身体的症状や精神的症状を呈する。これらの症状に対し,女性三大漢方薬の1つである加味逍遙散(KSS)が汎用され,特にうつ・不安,イライラなどの精神的症状改善において臨床的評価が高い。本研究は,更年期うつ症状を改善するKSSの作用機序解明を目的とし,更年期うつ病モデルマウスを用いた解析を行った結果,KSSが海馬におけるセロトニン1A(5-HT1A)受容体発現回復およびPKA-CREB-BDNF経路の活性化を介してうつ症状を改善している可能性が示唆された。
  • 清水 尚子; 石野 雄吾; 武田 卓; 遠山 正彌; 宮田 信吾
    産婦人科の実際 72 3 231 - 235 金原出版(株) 2023年03月 
    <文献概要>女性の更年期障害は,卵巣機能低下によるエストロゲン分泌減少などの生物学的要因に加え,社会心理的ストレスなどの環境要因が複雑に関与し,身体的症状や精神的症状を呈する。これらの症状に対し,女性三大漢方薬の1つである加味逍遙散(KSS)が汎用され,特にうつ・不安,イライラなどの精神的症状改善において臨床的評価が高い。本研究は,更年期うつ症状を改善するKSSの作用機序解明を目的とし,更年期うつ病モデルマウスを用いた解析を行った結果,KSSが海馬におけるセロトニン1A(5-HT1A)受容体発現回復およびPKA-CREB-BDNF経路の活性化を介してうつ症状を改善している可能性が示唆された。
  • Miyata S; Ishino Y; Shimizu S; Tohyama M
    Front Aging Neurosci. 14 934346 - 934346 2022年07月 [査読有り]
     
    Major depressive disorder (MDD) is a multifactorial disease affected by several environmental factors. Although several potential onset hypotheses have been identified, the molecular mechanisms underlying the pathogenesis of this disorder remain unclear. Several recent studies have suggested that among many environmental factors, inflammation and immune abnormalities in the brain or the peripheral tissues are associated with the onset of MDDs. Furthermore, several stress-related hypotheses have been proposed to explain the onset of MDDs. Thus, inflammation or immune abnormalities can be considered stress responses that occur within the brain or other tissues and are regarded as one of the mechanisms underlying the stress hypothesis of MDDs. Therefore, we introduce several current advances in inflammation studies in the brain that might be related to the pathophysiology of MDD due to stress exposure in this review.
  • Ishino Y; Shimizu S; Tohyama M; Miyata S
    Dev Neurobiol. 82 3 245 - 260 2022年04月 [査読有り]
     
    Protein arginine methylation has been recognized as one of key posttranslational modifications for refined protein functions, mediated by protein arginine methyltransferases (Prmts). Coactivator-associated arginine methyltransferase (Carm1, also known as Prmt4) participates in various cellular events, such as cell survival, proliferation, and differentiation through its protein arginine methylation activities. Carm1 regulates cell proliferation of a neuronal cell line and is reportedly expressed in the mammalian brain. However, its detailed function in the central nervous system, particularly in glial cells, remains largely unexplored. In this study, Carm1 exhibited relatively high expression in oligodendrocyte (OL) lineage cells present in the corpus callosum of the developing brain, followed by a remarkable downregulation after active myelination. The suppression of Carm1 activity by inhibitors in isolated oligodendrocyte precursor cells (OPCs) reduced the number of Ki67-expressing and BrdU-incorporated proliferating cells. Furthermore, Carm1 inactivation attenuated OL differentiation, as determined by the expression of Plp, a reliable myelin-related marker. It also impaired the extension of OL processes, accompanied by a significant reduction in gene expression related to OL differentiation and myelination, such as Sox10, Cnp, Myrf, and Mbp. In addition, OLs co-cultured with embryonic dorsal root ganglia neurons demonstrated that Carm1 activity is required for the appropriate formation of myelin processes and myelin sheaths around neuronal axons, and the induction of the clustering of Caspr, a node of Ranvier structural molecule. Thus, we propose that Carm1 is an essential molecule for the development of OPCs and OLs during brain development.
  • 更年期障害における精神神経症状を改善する加味逍遙散の作用機序解明
    清水 尚子; 石野 雄吾; 武田 卓; 遠山 正彌; 宮田 信吾
    日本自律神経学会総会プログラム・抄録集 74回 108 - 108 日本自律神経学会 2021年10月
  • Miyata S
    Myelin, Basic and Clinical Advances, Advances in Experimental Medicine and Biology 1190 33 - 42 2019年11月 [査読有り][招待有り]
  • うつ病とレム睡眠の関係について マウスのうつ病モデルを用いた解析
    安垣 進之助; 劉 至堯; 柏木 光昭; 鹿糠 実香; 本多 隆利; 宮田 信吾; 柳沢 正史; 林 悠
    日本睡眠学会定期学術集会プログラム・抄録集 44回 252 - 252 (一社)日本睡眠学会 2019年06月
  • Shoko Shimizu; Yugo Ishino; Takashi Takeda; Masaya Tohyama; Shingo Miyata
    Evidence-based complementary and alternative medicine : eCAM 2019 9475384 - 9475384 2019年 [査読有り]
     
    Females are well known to suffer disproportionately more than males from stress-related neuropsychiatric disorders, especially during perimenopausal and postmenopausal periods. In addition to a decline in serum estradiol levels, environmental stress and social stress likely contribute to the development of neuropsychiatric symptoms in perimenopausal and postmenopausal women. Kamishoyosan (KSS) is a traditional Japanese Kampo medicine, composed of a specified mixture of 10 crude compounds derived from plant sources, widely used for various neuropsychiatric symptoms in perimenopausal and postmenopausal women. However, the molecular mechanisms underlying KSS-mediated attenuation of neuropsychological symptoms and stress-response behaviors in perimenopausal and postmenopausal women remain unknown. In the present study, we first established a mouse model for postmenopausal depression-like signs using chronic water-immersion and restraint-stressed ovariectomized (OVX) mice to investigate the underlying molecular mechanism of KSS. We found that continuous administration of KSS to these mice normalized the activation of the hypothalamic-pituitary-adrenal (HPA) axis, ameliorated stress-induced depressive behavior, and prevented a decrease of neurogenesis in the hippocampus. As previous studies have implicated dysfunction of the hippocampal 5-HT1A receptor (5-HT1AR) in depressive disorders, we also evaluated the effect of KSS on 5-HT1AR expression and the protein kinase A- (PKA-) cAMP response element-binding- (CREB-) brain-derived neurotrophic factor (BDNF) signaling pathway in the hippocampus in this model. The level of 5-HT1AR in the hippocampus decreased in chronic stress-exposed OVX mice, while KSS treatment normalized the stress-induced decrease in 5-HT1AR expression in the hippocampus of chronic stress-exposed OVX mice. Furthermore, we found that KSS treatment upregulated the expression levels of phosphorylated PKA (p-PKA), phosphorylated CREB (p-CREB), and BDNF in the hippocampus in chronic stress-exposed OVX mice. These results suggest that KSS improves neuropsychiatric symptoms through 5-HT1AR and PKA-CREB-BDNF signaling in the hippocampus in postmenopausal women.
  • Shinnosuke Yasugaki; Chih-Yao Liu; Mitsuaki Kashiwagi; Mika Kanuka; Takato Honda; Shingo Miyata; Masashi Yanagisawa; Yu Hayashi
    Frontiers in neuroscience 13 1072 - 1072 2019年 [査読有り]
     
    Repeated stress is a risk factor for mental disorders and can also lead to sleep disturbances. Although the effects of stress on sleep architecture have been investigated in rodents, the length of the stress exposure period in most studies has been limited to about 10 days, and few studies have analyzed the effects of chronic stress over a longer period. Here we investigated how sleep is affected in a mouse model of depression induced by 3 weeks of daily water immersion and restraint stress (WIRS). Sleep was recorded after 1, 2, and 3 weeks of stress exposure. Some stress-induced changes in several sleep measures were maintained across the 3 weeks, whereas other changes were most prominent during the 1st week. The total amount of non-rapid eye movement sleep (NREMS) was increased and the total amount of time spent awake was decreased across all 3 weeks. On the other hand, the amount of REMS during the dark phase was significantly increased in the 1st week compared with that at baseline or the 2nd and 3rd weeks. Electroencephalogram (EEG) power in the delta range was decreased during NREMS, although the total amount of NREMS was increased. These findings indicate that repeated WIRS, which eventually leads to a depression-like phenotype, differentially affects sleep between the early and subsequent periods. The increase in the amount of REMS during the dark phase in the 1st week significantly correlated with changes in body weight. Our results show how sleep changes throughout a long period of chronic stress in a mouse model of depression.
  • Shoko Shimizu; Yugo Ishino; Masaya Tohyama; Shingo Miyata
    Scientific Reports 8 1 7644  2018年12月 [査読有り]
     
    Oligodendrocytes, the myelin-forming cells in the central nervous system (CNS), undergo morphological differentiation characterized by elaborated branched processes to enwrap neuronal axons. However, the basic molecular mechanisms underlying oligodendrocyte morphogenesis remain unknown. Herein, we describe the essential roles of Nuclear Distribution E Homolog 1 (NDE1), a dynein cofactor, in oligodendrocyte morphological differentiation. In the mouse corpus callosum, Nde1 mRNA expression was detected in oligodendrocyte lineage cells at the postnatal stage. In vitro analysis revealed that downregulation of NDE1 by siRNA impaired the outgrowth and extensive branching of oligodendrocyte processes and led to a decrease in the expression of myelin-related markers, namely, CNPase and MBP. In myelinating co-cultures with dorsal root ganglion (DRG) neurons, NDE1-knockdown oligodendrocyte precursor cells (OPCs) failed to develop into MBP-positive oligodendrocytes with multiple processes contacting DRG axons. Immunoprecipitation studies showed that NDE1 interacts with the dynein intermediate chain (DIC) in oligodendrocytes, and an overexpressed DIC-binding region of NDE1 exerted effects on oligodendrocyte morphogenesis that were similar to those following NDE1 knockdown. Furthermore, NDE1-knockdown-impaired oligodendrocyte process formation was rescued by siRNA-resistant wild-type NDE1 but not by DIC-binding region-deficient NDE1 overexpression. These results suggest that NDE1 plays a crucial role in oligodendrocyte morphological differentiation via interaction with dynein.
  • Tanaka T; Shimizu S; Ueno M; Fujihara Y; Ikawa M; Miyata S
    EBioMedicine 30 62 - 73 2018年04月 [査読有り]
     
    © 2018 The Authors Abnormalities in limbic neural circuits have been implicated in the onset of anxiety disorders. However, the molecular pathogenesis underlying anxiety disorders remains poorly elucidated. Here, we demonstrate that myristoylated alanine-rich C-kinase substrate like 1 (MARCKSL1) regulates amygdala circuitry to control the activity of the hypothalamic-pituitary-adrenal (HPA) axis, as well as induces anxiety-like behaviors in mice. MARCKSL1 expression was predominantly localized in the prefrontal cortex (PFC), hypothalamus, hippocampus, and amygdala of the adult mouse brain. MARCKSL1 transgenic (Tg) mice exhibited anxiety-like behaviors dependent on corticotropin-releasing hormone. MARCKSL1 increased spine formation in the central amygdala, and downregulation of MARCKSL1 in the amygdala normalized both increased HPA axis activity and elevated anxiety-like behaviors in Tg mice. Furthermore, MARCKSL1 expression was increased in the PFC and amygdala in a brain injury model associated with anxiety-like behaviors. Our findings suggest that MARCKSL1 expression in the amygdala plays an important role in anxiety-like behaviors.
  • Shingo Miyata; Manabu Taniguchi; Yoshihisa Koyama; Shoko Shimizu; Takashi Tanaka; Fumihiko Yasuno; Akihide Yamamoto; Hidehiro Iida; Takashi Kudo; Taiichi Katayama; Masaya Tohyama
    SCIENTIFIC REPORTS 6 23084  2016年03月 [査読有り]
     
    Repeated stressful events are associated with the onset of major depressive disorder (MDD). We previously showed oligodendrocyte (OL)-specific activation of the serum/glucocorticoid-regulated kinase (SGK)1 cascade, increased expression of axon-myelin adhesion molecules, and elaboration of the oligodendrocytic arbor in the corpus callosum of chronically stressed mice. In the current study, we demonstrate that the nodes and paranodes of Ranvier in the corpus callosum were narrower in these mice. Chronic stress also led to diffuse redistribution of Caspr and Kv 1.1 and decreased the activity in white matter, suggesting a link between morphological changes in OLs and inhibition of axonal activity. OL primary cultures subjected to chronic stress resulted in SGK1 activation and translocation to the nucleus, where it inhibited the transcription of metabotropic glutamate receptors (mGluRs). Furthermore, the cAMP level and membrane potential of OLs were reduced by chronic stress exposure. We showed by diffusion tensor imaging that the corpus callosum of patients with MDD exhibited reduced fractional anisotropy, reflecting compromised white matter integrity possibly caused by axonal damage. Our findings suggest that chronic stress disrupts the organization of the nodes of Ranvier by suppressing mGluR activation in OLs, and that specific white matter abnormalities are closely associated with MDD onset.
  • Shingo Miyata; Keiko Yoshikawa; Manabu Taniguchi; Toshiko Ishikawa; Takashi Tanaka; Shoko Shimizu; Masaya Tohyama
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 464 1 76 - 82 2015年08月 [査読有り]
     
    Major depression, one of the most prevalent mental illnesses, is thought to be a multifactorial disease related to both genetic and environmental factors. However, the genes responsible for and the pathogenesis of major depression at the molecular level remain unclear. Recently, we reported that stressed mice with elevated plasma corticosterone levels show upregulation and activation of serum glucocorticoid-regulated kinase (Sgk1) in oligodendrocytes. Active Sgk1 causes phosphorylation of N-myc downstream-regulated gene 1 (Ndrg1), and phospho-Ndrg1 increases the expression of N-cadherin, alpha-catenin, and beta-catenin in oligodendrocytes. This activation of the Sgk1 cascade results in morphological changes in the oligodendrocytes of nerve fiber bundles, such as those present in the corpus callosum. However, little is known about the molecular functions of the traditional and/or desmosomal cadherin superfamily in oligodendrocytes. Therefore, in this study, we aimed to elucidate the functions of the desmosomal cadherin superfamily in oligodendrocytes. Desmoglein (Dsg) 1, Dsg2, and desmocollin 1 (Dsc1) were found to be expressed in the corpus callosum of mouse brain, and the expression of a subtype of Dsg1, Dsg1c, was upregulated in oligodendrocytes after chronic stress exposure. Furthermore, Dsg1 proteins were localized around the plasma membrane regions of oligodendrocytes. A study in primary oligodendrocyte cultures also revealed that chronic upregulation of Sgk1 by dexamethasone administration is involved in upregulation of Dsg1c mRNA. These results may indicate that chronic stress induced Sgk1 activation in oligodendrocytes, which increases Dsg1 expression near the plasma membrane. Thus, Dsg1 upregulation may be implicated in the molecular mechanisms underlying the morphological changes in oligodendrocytes in response to chronic stress exposure. (C) 2015 Elsevier Inc. All rights reserved.
  • Masaya Tohyama; Shingo Miyata; Tsuyoshi Hattori; Shoko Shimizu; Shinsuke Matsuzaki
    ANATOMICAL SCIENCE INTERNATIONAL 90 3 137 - 143 2015年06月 [査読有り]
     
    Recently several potential susceptibility genes for major psychiatric disorders (schizophrenia and major depression) such as disrupted-in-schizophrenia 1(DISC1), dysbindin and pituitary adenylate cyclase-activating polypeptide (PACAP) have been reported. DISC1 is involved in neural development directly via adhesion molecules or via its binding partners of DISC1 such as elongation protein zeta-1 (FEZ1), DISC1-binding zinc-finger protein (DBZ) and kendrin. PACAP also regulates neural development via stathmin 1 or via regulation of the DISC1-DBZ binding. Dysbindin is also involved in neural development by regulating centrosomal microtubule network formation. All such molecules examined to date are involved in neural development. Thus, these findings provide new molecular insights into the mechanisms of neural development and neuropsychiatric disorders. On the other hand, in addition to neurons, both DISC and DBZ have been detected in oligodendrocytes and implicated in regulating oligodendrocyte differentiation. DISC1 inhibits the differentiation of oligodendrocyte precursor cells into oligodendrocytes, while DBZ has a positive regulatory role in oligodendrocyte differentiation. Evidence suggesting that disturbance of oligodendrocyte development causes major depression is also described.
  • Shoko Shimizu; Takashi Tanaka; Masaya Tohyama; Shingo Miyata
    BRAIN RESEARCH BULLETIN 114 49 - 55 2015年05月 [査読有り]
     
    Stressful events are known to down-regulate expression levels of glucocorticoid receptors (GRs) in the brain. Recently, we reported that stressed mice with elevated plasma levels of corticosterone exhibit morphological changes in the oligodendrocytes of nerve fiber bundles, such as those in the corpus callosum. However, little is known about the molecular mechanism of GR expression regulation in oligodendrocytes after stress exposure. A previous report has suggested that GR protein levels might be regulated by microRNA (miR)-18 and/or -124a in the brain. In this study, we aimed to elucidate the GR regulation mechanism in oligodendrocytes and evaluate the effects of yokukansan (YKS), a Kampo medicine, on GR protein regulation. Acute exposure to stress increased plasma corticosterone levels, decreased GR protein expression, and increased miR-124a expression in the corpus callosum of adult male mice, though the GR mRNA and miR-18 expression levels were not significant changes. YKS normalized the stress-induced changes in the plasma corticosterone, GR protein, and miR124a expression levels. An oligodendrocyte primary culture study also showed that YKS down-regulated miR-124a, but not miR-18, expression levels in dexamethasone-treated cells. These results suggest that the down-regulation of miR124a expression might be involved in the normalization of stress-induced decreases in GR protein in oligodendrocytes by YKS. This effect may imply the molecular mechanisms underlying the ameliorative effects of YKS on psychological symptoms and stress-related behaviors. (C) 2015 Elsevier Inc. All rights reserved.
  • Shingo Miyata; Tsuyoshi Hattori; Shoko Shimizu; Akira Ito; Masaya Tohyama
    BIOMED RESEARCH INTERNATIONAL 2015 492367  2015年 [査読有り]
     
    The major psychiatric disorders such as schizophrenia (SZ) and major depressive disorder (MDD) are thought to be multifactorial diseases related to both genetic and environmental factors. However, the genes responsible and the molecular mechanisms underlying the pathogenesis of SZ and MDD remain unclear. We previously reported that abnormalities of disrupted-in-Schizophrenia-1 (DISC1) and DISC1 binding zinc finger (DBZ) might cause major psychiatric disorders such as SZ. Interestingly, both DISC and DBZ have been further detected in oligodendrocytes and implicated in regulating oligodendrocyte differentiation. DISC1 negatively regulates the differentiation of oligodendrocytes, whereas DBZ plays a positive regulatory role in oligodendrocyte differentiation. We have reported that repeated stressful events, one of the major risk factors of MDD, can induce sustained upregulation of plasma corticosterone levels and serum/glucocorticoid regulated kinase 1 (Sgk1) mRNA expression in oligodendrocytes. Repeated stressful events can also activate the SGK1 cascade and cause excess arborization of oligodendrocyte processes, which is thought to be related to depressive-like symptoms. In this review, we discuss the expression of DISC1, DBZ, and SGK1 in oligodendrocytes, their roles in the regulation of oligodendrocyte function, possible interactions of DISC1 and DBZ in relation to SZ, and the activation of the SGK1 signaling cascade in relation to MDD.
  • Shoko Shimizu; Takashi Tanaka; Takashi Takeda; Masaya Tohyama; Shingo Miyata
    BIOMED RESEARCH INTERNATIONAL 2015 797280  2015年 [査読有り]
     
    It is well known that glucocorticoid receptor (GR) signaling regulates the hypothalamic-pituitary-adrenal (HPA) axis, and GR expression level is associated with HPA axis activity. Recent studies revealed that microRNA-(miR-) 18 and/or 124a are candidate negative regulators of GR in the brain. The Kampo medicine Yokukansan (YKS) can affect psychological symptoms such as depression and anxiety that are associated with stress responses. In this study, we evaluated the effect of YKS on miR-18 and 124a and GR levels in mice exposed to stress. We found that YKS pretreatment normalized elevated plasma corticosterone levels in stress-exposed mice. In addition, GR mRNA levels were downregulated in the brain following stress exposure. While miR-124a expression levels were not altered in the hypothalamus of stress-exposed mice, miR-18 levels decreased in the hypothalamus of YKS-pretreated mice after stress exposure. Finally, GR protein levels in the paraventricular nucleus (PVN) of the hypothalamus after stress exposure recovered in YKS-pretreated mice. Collectively, these data suggest that YKS normalizes GR protein levels by regulating miR-18 expression in the hypothalamus, thus normalizing HPA axis activity following stress exposure.
  • Shoko Shimizu; Yoshihisa Koyama; Tsuyoshi Hattori; Taro Tachibana; Tomohiko Yoshimi; Hisayo Emoto; Yuji Matsumoto; Shingo Miyata; Taiichi Katayama; Akira Ito; Masaya Tohyama
    GLIA 62 5 709 - 724 2014年05月 [査読有り]
     
    Recent studies have shown changes in myelin genes and alterations in white matter structure in a wide range of psychiatric disorders. Here we report that DBZ, a central nervous system (CNS)-specific member of the DISC1 interactome, positively regulates the oligodendrocyte (OL) differentiation in vivo and in vitro. In mouse corpus callosum (CC), DBZ mRNA is expressed in OL lineage cells and expression of DBZ protein peaked before MBP expression. In the CC of DBZ-KO mice, we observed delayed myelination during the early postnatal period. Although the myelination delay was mostly recovered by adulthood, OLs with immature structural features were more abundant in adult DBZ-KO mice than in control mice. DBZ was also transiently upregulated during rat OL differentiation in vitro before myelin marker expression. DBZ knockdown by RNA interference resulted in a decreased expression of myelin-related markers and a low number of cells with mature characteristics, but with no effect on the proliferation of oligodendrocyte precursor cells. We also show that the expression levels of transcription factors having a negative-regulatory role in OL differentiation were upregulated when endogenous DBZ was knocked down. These results strongly indicate that OL differentiation in rodents is regulated by DBZ.
  • Tomoko Tanigawa; Shigeyuki Kanazawa; Ryoko Ichibori; Takashi Fujiwara; Takuya Magome; Kenta Shingaki; Shingo Miyata; Yuki Hata; Koichi Tomita; Ken Matsuda; Tateki Kubo; Masaya Tohyama; Kenji Yano; Ko Hosokawa
    BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 14 133  2014年04月 [査読有り]
     
    Background: Oxidative stress has been suggested as a mechanism underlying skin aging, as it triggers apoptosis in various cell types, including fibroblasts, which play important roles in the preservation of healthy, youthful skin. Catechins, which are antioxidants contained in green tea, exert various actions such as anti-inflammatory, anti-bacterial, and anti-cancer actions. In this study, we investigated the effect of (+)-catechin on apoptosis induced by oxidative stress in fibroblasts. Methods: Fibroblasts (NIH3T3) under oxidative stress induced by hydrogen peroxide (0.1 mM) were treated with either vehicle or (+)-catechin (0-100 mu M). The effect of (+)-catechin on cell viability, apoptosis, phosphorylation of c-Jun terminal kinases (JNK) and p38, and activation of caspase-3 in fibroblasts under oxidative stress were evaluated. Results: Hydrogen peroxide induced apoptotic cell death in fibroblasts, accompanied by induction of phosphorylation of JNK and p38 and activation of caspase-3. Pretreatment of the fibroblasts with (+)-catechin inhibited hydrogen peroxide-induced apoptosis and reduced phosphorylation of JNK and p38 and activation of caspase-3. Conclusion: (+)-Catechin protects against oxidative stress-induced cell death in fibroblasts, possibly by inhibiting phosphorylation of p38 and JNK. These results suggest that (+)-catechin has potential as a therapeutic agent for the prevention of skin aging.
  • Takashi Fujiwara; Shigeyuki Kanazawa; Ryoko Ichibori; Tomoko Tanigawa; Takuya Magome; Kenta Shingaki; Shingo Miyata; Masaya Tohyama; Ko Hosokawa
    PLOS ONE 9 3 e92168  2014年03月 [査読有り]
     
    L-Arginine is considered a conditionally essential amino acid and has been shown to enhance wound healing. However, the molecular mechanisms through which arginine stimulates cutaneous wound repair remain unknown. Here, we evaluated the effects of arginine supplementation on fibroblast proliferation, which is a key process required for new tissue formation. We also sought to elucidate the signaling pathways involved in mediating the effects of arginine on fibroblasts by evaluation of extracellular signal-related kinase (ERK) 1/2 activation, which is important for cell growth, survival, and differentiation. Our data demonstrated that addition of 6 mM arginine significantly enhanced fibroblast proliferation, while arginine deprivation increased apoptosis, as observed by enhanced DNA fragmentation. In vitro kinase assays demonstrated that arginine supplementation activated ERK1/2, Akt, PKA and its downstream target, cAMP response element binding protein (CREB). Moreover, knockdown of GPRC6A using siRNA blocked fibroblast proliferation and decreased phosphorylation of ERK1/2, Akt and CREB. The present experiments demonstrated a critical role for the GPRC6A-ERK1/2 and PI3K/Akt signaling pathway in arginine-mediated fibroblast survival. Our findings provide novel mechanistic insights into the positive effects of arginine on wound healing.
  • Tameyasu Maeda; Manabu Taniguchi; Kenta Shingaki; Shigeyuki Kanazawa; Shingo Miyata
    ACUPUNCTURE IN MEDICINE 32 1 90 - 92 2014年02月 [査読有り]
  • Ayaka Ikeda; Shingo Miyata; Akihito Yokosuka; Yoshihiro Mimaki; Yasushi Ohizumi; Masakuni Degawa; Kiyomitsu Nemoto
    Fundamental Toxicological Sciences 1 4 169 - 172 2014年
  • Takuya Magome; Tsuyoshi Hattori; Manabu Taniguchi; Toshiko Ishikawa; Shingo Miyata; Kohei Yamada; Hironori Takamura; Shinsuke Matsuzaki; Akira Ito; Masaya Tohyama; Taiichi Katayama
    NEUROCHEMISTRY INTERNATIONAL 63 6 561 - 569 2013年11月 [査読有り]
     
    X-linked mental retardation (XLMR) is a common cause of moderate to severe intellectual disability in males. XLMR protein related to neurite extension (Xpn, also known as KIAA2022) has been implicated as a gene responsible for XLMR in humans. Although Xpn is highly expressed in the developing brain and is involved in neurite outgrowth in PC12 cells and neurons, little is known about the functional role of Xpn. Here, we show that Xpn regulates cell-cell and cell-matrix adhesion and migration in PC12 cells. Xpn knockdown enhanced cell-cell and cell-matrix adhesion mediated by N-cadherin and beta 1-integrin, respectively. N-Cadherin and beta 1-integrin expression at the mRNA and protein levels was significantly increased in Xpn knockdown PC12 cells. Furthermore, overexpressed Xpn protein was strongly expressed in the nuclei of PC12 and 293T cells. Finally, depletion of Xpn perturbed cellular migration by enhancing N-cadherin and beta 1-integrin expression in a PC12 cell wound healing assay. We conclude that Xpn regulates cell-cell and cell-matrix adhesion and cellular migration by regulating the expression of adhesion molecules. (C) 2013 Elsevier Ltd. All rights reserved.
  • Ayaka Ikeda; Kiyomitsu Nemoto; Chiaki Yoshida; Shingo Miyata; Junki Mori; Saori Soejima; Akihito Yokosuka; Yoshihiro Mimaki; Yasushi Ohizumi; Masakuni Degawa
    NEUROSCIENCE LETTERS 549 135 - 139 2013年08月 [査読有り]
     
    Increased expression of thioredoxin-interacting protein (TXNIP) has recently been proved to be a crucial event for irremediable endoplasmic reticulum (ER) stress resulting in the programmed cell death (apoptosis) of pancreatic beta-cells. The present study demonstrated that treatment with 1-10 mu g/ml tunicamycin, a potent revulsant of ER stress, drastically induced TXNIP expression accompanied by the generation of cleaved caspase-3 as an indicator of apoptosis in SK-N-SH human neuroblastoma cells. This result substantiated that TXNIP is also involved in neurodegeneration triggered by ER stress. Moreover, we evaluated the effects of nobiletin, a citrus polymethoxyflavonoid, on tunicamycin-induced apoptosis and TXNIP expression in SK-N-SH cells, because we reported previously that this flavonoid might be able to reduce TXNIP expression. Co-treatment of SK-N-SH cells with 100 mu M nobiletin and 1 mu g/ml tunicamycin for 24 h strongly suppressed apoptosis and increased TXNIP expression induced by 1 mu g/ml tunicamycin treatment alone. In addition, we proved that the ability of 100 mu M nobiletin treatment to reduce TXNIP expression is exerted from 3 h after the onset of treatment. Therefore, the protective and ameliorative effects of nobiletin on neuronal degeneration and impaired memory, which several studies using animal models have demonstrated, might arise in part from nobiletin's ability to repress TXNIP expression (C) 2013 Elsevier Ireland Ltd. All rights reserved.
  • Shingo Miyata; Tatsunori Mizuno; Yoshihisa Koyama; Taiichi Katayama; Masaya Tohyama
    PLOS ONE 8 7 e69732  2013年07月 [査読有り]
     
    The Golgi apparatus is important for the transport of secretory cargo. Glycosylation is a major post-translational event. Recognition of O-glycans on proteins is necessary for glycoprotein trafficking. In this study, specific inhibition of O-glycosylation (Golgi stress) induced the expression of endoplasmic reticulum (ER)-resident heat shock protein (HSP) 47 in NIH3T3 cells, although cell death was not induced by Golgi stress alone. When HSP47 expression was downregulated by siRNA, inhibition of O-glycosylation caused cell death. Three days after the induction of Golgi stress, the Golgi apparatus was disassembled, many vacuoles appeared near the Golgi apparatus and extended into the cytoplasm, the nuclei had split, and cell death assay-positive cells appeared. Six hours after the induction of Golgi stress, HSP47-knockdown cells exhibited increased cleavage of Golgi-resident caspase-2. Furthermore, activation of mitochondrial caspase-9 and ER-resident unfolded protein response (UPR)-related molecules and efflux of cytochrome c from the mitochondria to the cytoplasm was observed in HSP47-knockdown cells 24 h after the induction of Golgi stress. These findings indicate that (i) the ER-resident chaperon HSP47 protected cells from Golgi stress, and (ii) Golgi stress-induced cell death caused by the inhibition of HSP47 expression resulted from caspase-2 activation in the Golgi apparatus, extending to the ER and mitochondria.
  • Tameyasu Maeda; Manabu Taniguchi; Shinsuke Matsuzaki; Kenta Shingaki; Shigeyuki Kanazawa; Shingo Miyata
    ACUPUNCTURE IN MEDICINE 31 1 117 - 119 2013年03月 [査読有り]
  • Hironori Takamura; Yoshihisa Koyama; Shinsuke Matsuzaki; Kohei Yamada; Tsuyoshi Hattori; Shingo Miyata; Kana Takemoto; Masaya Tohyama; Taiichi Katayama
    PLOS ONE 7 12 e51912  2012年12月 [査読有り]
     
    Mitochondria are dynamic organelles that change in response to extracellular stimuli. These changes are essential for normal mitochondrial/cellular function and are controlled by a tight balance between two antagonistic pathways that promote fusion and fission. Although some molecules have been identified to mediate the mitochondrial fusion and fission process, the underlying mechanisms remain unclear. Tumor necrosis factor receptor-associated protein 1 (TRAP1) is a mitochondrial molecule that regulates a variety of mitochondrial functions. Here, we examined the role of TRAP1 in the regulation of morphology. Stable TRAP1 knockdown cells showed abnormal mitochondrial morphology, and we observed significant decreases in dynamin-related protein 1 (Drp1) and mitochondrial fission factor (Mff), mitochondrial fission proteins. Similar results were obtained by transient knockdown of TRAP1 in two different cell lines, SH-SY5Y neuroblastoma cells and KNS-42 glioma cells. However, TRAP1 knockdown did not affect expression levels of fusion proteins. The reduction in Drp1 and Mff protein levels was rescued following treatment with the proteasome inhibitor MG132. These results suggest that TRAP1 regulates the expression of fission proteins and controls mitochondrial fusion/fission, which affects mitochondrial/cellular function.
  • Kenta Shingaki; Manabu Taniguchi; Shigeyuki Kanazawa; Shinsuke Matsuzaki; Tameyasu Maeda; Shingo Miyata; Tateki Kubo; Kosuke Torii; Sadao Shiosaka; Masaya Tohyama
    JOURNAL OF DERMATOLOGICAL SCIENCE 67 1 71 - 73 2012年07月 [査読有り]
  • T. Ishikawa; S. Miyata; Y. Koyama; K. Yoshikawa; T. Hattori; N. Kumamoto; K. Shingaki; T. Katayama; M. Tohyama
    NEUROSCIENCE 214 181 - 191 2012年07月 [査読有り]
     
    KIAA2022 has been implicated as a gene responsible for expressing X-linked mental retardation (XLMR) proteins in humans. However, the functional role of KIAA2022 in the human brain remains unclear. Here, we revealed that depletion of Kiaa2022 inhibits neurite outgrowth of PC12 cells, indicating that the gene participates in neurite extension. Thus, we termed Kiaa2022 as an XLMR protein related to neurite extension (Xpn). Using the mouse brain as a model and ontogenetic analysis of Xpn by real-time PCR, we clearly demonstrated that Xpn is expressed transiently during the late embryonic and perinatal stages. In situ hybridization histochemistry further revealed that Xpn-expressing neurons could be categorized ontogenetically into three types. The first type showed transient expression of Xpn during development. The second type maximally expressed Xpn during the late embryonic or perinatal stage. Thereafter, Xpn expression in this type of neuron decreased gradually throughout development. Nevertheless, a significant level of Xpn expression was detected even into adulthood. The third type of neurons initiated expression of Xpn during the embryonic stage, and continued to express the gene throughout the remaining developmental stages. Subsequent immunohistochemical analysis revealed that Xpn was localized to the nucleus and cytoplasm throughout brain development. Our findings indicate that Xpn may participate in neural circuit formation during developmental stages via nuclear and cytoplasmic Xpn. Moreover, disturbances of this neuronal circuit formation may play a role in the pathogenesis of mental retardation. (c) 2012 IBRO. Published by Elsevier Ltd. All rights reserved.
  • Psychosis and Adhesion Molecules.
    Hattori T; Miyata S; Ito A; Katayama T; Tohyama M
    InTech, Mental Illness Book 1 2012年 [査読有り]
  • オリゴデンドロサイト特異的なシグナル伝達機構とうつ病発症との関連(Analysis of the intercellular signaling cascade induced by chronic stress exposurein oligodendrocytes in corpus callosum)
    宮田 信吾; 小山 佳久; 谷口 学; 吉川 景子; 石川 淑子; 弓場 智雄; 遠山 正彌
    神経化学 50 2-3 166 - 166 2011年09月
  • 自閉症関連因子Marcksl1の神経ネットワーク形成機構への関与(Involvement of autism-related factor MARCKSL1 in Neural network formation)
    石川 淑子; 宮田 信吾; 小山 佳久; 松崎 伸介; 片山 泰一; 遠山 正彌
    神経化学 50 2-3 185 - 185 2011年09月
  • ミトコンドリア形態制御におけるTRAP1の役割(Involvement of TRAP1 inregulation of mitochondrial morphology)
    高村 明孝; 宮田 信吾; 嶽本 香菜; 小山 佳久; 松崎 伸介; 遠山 正彌; 片山 泰一
    神経化学 50 2-3 188 - 188 2011年09月
  • Kana Takemoto; Shingo Miyata; Hironori Takamura; Taiichi Katayama; Masaya Tohyama
    NEUROCHEMISTRY INTERNATIONAL 58 8 880 - 887 2011年07月 [査読有り]
     
    Stress in mitochondria or the endoplasmic reticulum (ER) independently causes cell death. Recently, it was reported that ER stress causes mitochondrial dysfunction via p53-upregulated modulator of apoptosis (PUMA). However, little is known regarding the mitochondria molecules that mediate ER dysfunction. The present study revealed that tumor necrosis factor receptor-associated protein 1 (TRAP1), which localizes in the mitochondria, is associated with the unfolded protein response (UPR) in the ER. TRAP1 knockdown activated the ER-resident caspase-4, which is activated by ER stress, to induce cell death in humans. However, TRAP1 knockdown cells did not show a significant increase in the level of cell death at least within 24 h after early phase of ER stress in comparison with that of the control cells. This finding could be attributed to a number of reasons. TRAP1 knockdown failed to activate caspase-9, which is activated by activated caspase-4. In addition, TRAP1 knockdown increased the basal level of GRP78/BiP expression, which protects cells, and decreased the basal level of C/EBP homologous protein (CHOP) expression, which induces cell death, even under ER stress. Thus, the present study revealed that mitochondria could be a potential regulator of the UPR in the ER through mitochondrial TRAP1. (C) 2011 Elsevier Ltd. All rights reserved.
  • Shingo Miyata; Yoshihisa Koyama; Kana Takemoto; Keiko Yoshikawa; Toshiko Ishikawa; Manabu Taniguchi; Kiyoshi Inoue; Miwa Aoki; Osamu Hori; Taiichi Katayama; Masaya Tohyama
    PLOS ONE 6 5 e19859  2011年05月 [査読有り]
     
    Repeated stressful events are known to be associated with onset of depression. Further, stress activates the hypothalamic-pituitary-adrenocortical (HPA) system by elevating plasma cortisol levels. However, little is known about the related downstream molecular pathway. In this study, by using repeated water-immersion and restraint stress (WIRS) as a stressor for mice, we attempted to elucidate the molecular pathway induced by elevated plasma corticosterone levels. We observed the following effects both, in vivo and in vitro: (1) repeated exposure to WIRS activates the 3-phosphoinositide-dependent protein kinase (PDK1)-serum glucocorticoid regulated kinase (SGK1)-N-myc downstream-regulated gene 1 (NDRG1)-adhesion molecule (i.e., N-cadherin, alpha-catenin, and beta-catenin) stabilization pathway via an increase in plasma corticosterone levels; (2) the activation of this signaling pathway induces morphological changes in oligodendrocytes; and (3) after recovery from chronic stress, the abnormal arborization of oligodendrocytes and depression-like symptoms return to the control levels. Our data strongly suggest that these abnomalities of oligodendrocytes are possibly related to depression-like symptoms.
  • Toru Hiratsuka; Shinsuke Matsuzaki; Shingo Miyata; Mitsuhiro Kinoshita; Kazuaki Kakehi; Shinji Nishida; Taiichi Katayama; Masaya Tohyama
    PLOS ONE 5 10 e13280  2010年10月 [査読有り]
     
    Background: Recently, several studies have reported Yokukansan (Tsumura TJ-54), a traditional Japanese medicine, as a potential new drug for the treatment of Alzheimer's disease (AD). Endoplasmic reticulum (ER) stress is known to play an important role in the pathogenesis of AD, particularly in neuronal death. Therefore, we examined the effect of Yokukansan on ER stress-induced neurotoxicity and on familial AD-linked presenilin-1 mutation-associated cell death. Methods: We employed the WST-1 assay and monitored morphological changes to evaluate cell viability following Yokukansan treatment or treatment with its components. Western blotting and PCR were used to observe the expression levels of GRP78/BiP, caspase-4 and C/EBP homologous protein. Results: Yokukansan inhibited neuronal death during ER stress, with Cnidii Rhizoma (Senkyu), a component of Yokukansan, being particularly effective. We also showed that Yokukansan and Senkyu affect the unfolded protein response following ER stress and that these drugs inhibit the activation of caspase-4, resulting in the inhibition of ER stress-induced neuronal death. Furthermore, we found that the protective effect of Yokukansan and Senkyu against ER stress could be attributed to the ferulic acid content of these two drugs. Conclusions: Our results indicate that Yokukansan, Senkyu and ferulic acid are protective against ER stress-induced neuronal cell death and may provide a possible new treatment for AD.
  • Shingo Miyata; Yasutake Mori; Masaya Tohyama
    BRAIN RESEARCH 1352 11 - 20 2010年09月 [査読有り]
     
    Protein arginine N-methyltransferase 3 (PRMT3) is a cytoplasmic enzyme that utilizes S-adenosyl-L-methionine (AdoMet) to methylate specific proteins, most of which contain GAR (glycine-arginine rich) motifs. PRMT3 has been shown to play a role in the proper maturation of the 80S ribosome by binding to and catalyzing the methylation of rpS2, a component of the 40S ribosomal subunit. However, the other roles of PRMT3 are fairly unclear, particularly in the brain, which is abundant in methylated proteins. In this study, we perturbed PRMT3 expression in cultured rat hippocampal neurons by transiently introducing siRNA oligonucleotides that were designed to hybridize with PRMT3 mRNA and then we examined the morphological and functional effects of neuronal PRMT3 depletion. PRMT3-defective neurons showed deformed spines without any change in spine number; less BDNF-induced protein translation of alpha CaMKII; and diminished rpS2 protein stability. Furthermore, overexpression of a methylation-resistant rpS2, whose methylated arginine residues were deleted, produced phenotypes that were similar to those associated with PRMT3 downregulation. These findings demonstrated that PRMT3 possibly plays a pivotal role in neuronal translation by interaction with rpS2 and that it contributes to activity-dependent changes in the dendritic spines. (C) 2010 Published by Elsevier B.V.
  • Shigeyuki Kanazawa; Toshihiro Fujiwara; Shinsuke Matsuzaki; Kenta Shingaki; Manabu Taniguchi; Shingo Miyata; Masaya Tohyama; Yasuo Sakai; Kenji Yano; Ko Hosokawa; Tateki Kubo
    PLOS ONE 5 8 e12228  2010年08月 [査読有り]
     
    Fibroblast proliferation and migration play important roles in wound healing. bFGF is known to promote both fibroblast proliferation and migration during the process of wound healing. However, the signal transduction of bFGF-induced fibroblast migration is still unclear, because bFGF can affect both proliferation and migration. Herein, we investigated the effect of bFGF on fibroblast migration regardless of its effect on fibroblast proliferation. We noticed involvement of the small GTPases of the Rho family, PI3-kinase, and JNK. bFGF activated RhoA, Rac1, PI3-kinase, and JNK in cultured fibroblasts. Inhibition of RhoA did not block bFGF-induced fibroblast migration, whereas inhibition of Rac1, PI3-kinase, or JNK blocked the fibroblastmigration significantly. PI3-kinase- inhibited cells down-regulated the activities of Rac1 and JNK, and Rac1-inhibited cells down-regulated JNK activity, suggesting that PI3-kinase is upstream of Rac1 and that JNK is downstream of Rac1. Thus, we concluded that PI3-kinase, Rac1, and JNK were essential for bFGF-induced fibroblast migration, which is a novel pathway of bFGF-induced cell migration.
  • ヒト脳におけるストレス関連因子SGK1の新規相互作用因子の同定と機能解析(Identification and characterization of novel interacting proteins of the stress-related factor SGK1 in the human brain)
    石川 淑子; 宮田 信吾; 吉川 景子; 小山 佳久; 遠山 正彌
    神経化学 49 2-3 737 - 737 2010年08月
  • Hiroaki Okuda; Ryusuke Kuwahara; Shinsuke Matsuzaki; Shingo Miyata; Natsuko Kumamoto; Tsuyoshi Hattori; Shoko Shimizu; Kohei Yamada; Keisuke Kawamoto; Ryota Hashimoto; Masatoshi Takeda; Taiichi Katayama; Masaya Tohyama
    PLOS ONE 5 1 e8773  2010年01月 [査読有り]
     
    Background: An accumulating body of evidence suggests that Dtnbp1 (Dysbindin) is a key susceptibility gene for schizophrenia. Using the yeast-two-hybrid screening system, we examined the candidate proteins interacting with Dysbindin and revealed one of these candidates to be the transcription factor NF-YB. Methods: We employed an immunoprecipitation (IP) assay to demonstrate the Dysbindin-NF-YB interaction. DNA chips were used to screen for altered expression of genes in cells in which Dysbindin or NF-YB was down regulated, while Chromatin IP and Reporter assays were used to confirm the involvement of these genes in transcription of Myristoylated alanine-rich protein kinase C substrate (MARCKS). The sdy mutant mice with a deletion in Dysbindin, which exhibit behavioral abnormalities, and wild-type DBA2J mice were used to investigate MARCKS expression. Results: We revealed an interaction between Dysbindin and NF-YB. DNA chips showed that MARCKS expression was increased in both Dysbindin knockdown cells and NF-YB knockdown cells, and Chromatin IP revealed interaction of these proteins at the MARCKS promoter region. Reporter assay results suggested functional involvement of the interaction between Dysbindin and NF-YB in MARCKS transcription levels, via the CCAAT motif which is a NF-YB binding sequence. MARCKS expression was increased in sdy mutant mice when compared to wild-type mice. Conclusions: These findings suggest that abnormal expression of MARCKS via dysfunction of Dysbindin might cause impairment of neural transmission and abnormal synaptogenesis. Our results should provide new insights into the mechanisms of neuronal development and the pathogenesis of schizophrenia.
  • A. Kousaka; Y. Mori; Y. Koyama; T. Taneda; S. Miyata; M. Tohyama
    NEUROSCIENCE 163 4 1146 - 1157 2009年11月 [査読有り]
     
    Protein arginine N-methyltransferase (PRMT) 8 was first discovered from a database search for genes harboring four conserved methyltransferase motifs, which shares more than 80% homology to PRMT1 in amino acid [Lee J, Sayegh J, Daniel J, Clarke S, Bedford MT (2005) PRMT8, a new membrane-bound tissue-specific member of the protein arginine methyltransferase family. J Biol Chem 280:32890-32896]. Interestingly, its tissue distribution is strikingly restricted to mouse CNS. To characterize the function in the CNS neurons, we raised an antiserum against PRMT8 to perform immunohistochemistry (IHC) and Western blot analysis. By IHC, the immunoreactivity of endogenous PRMT8 was broadly distributed in the CNS neurons with markedly intense signals in the cerebellum, hippocampal formation, and cortex, but was not detected in the cerebellar granular layer. In some subset of the neurons, the immunoreactivity was observed in the dendrites and axon bundles. The subcellular localization of the immunoreactivity was dominantly nuclear, arguing against the original report that exogenously expressed PRMT8 localizes to the plasma membrane via the N-terminal myristoylation. A series of the exogenously expressed proteins with different in-frame translation initiation codons was tested for comparison with the endogenous protein in molecular size. The third initiator codon produced the protein that was equivalent in size to the endogenous and showed a similar localizing pattern in PC12 cells. In conclusion, PRMT8 is a neuron-specific nuclear enzyme and the N-terminus does not contain the glycine end for myristoylation target. (C) 2009 IBRO. Published by Elsevier Ltd. All rights reserved.
  • Shingo Miyata; Yasutake Mori; Masaya Tohyama
    NEUROSCIENCE LETTERS 445 2 162 - 165 2008年11月 [査読有り]
     
    Neurite outgrowth is one of the Crucial events in the formation of neural circuits. The majority of studies on neurite outgrowth have focused on signal transduction processes based on phosphorylation and acetylation: a few studies have suggested the involvement of other molecular mechanisms. Recent progress in understanding the nature of protein arginine N-methyltransferases (PRMTs) raises the possibility of the involvement of protein methylation accompanied by cell shape changes during neuronal differentiation. Here, we show that PRMT1 play a pivotal role in the neurite outgrowth of Neuro2a cells. Our results revealed that PRMT1 depletion specifically affected neurite outgrowth but not the physiological processes involved in cell growth and differentiation. Furthermore, we demonstrated that Btg2, one of the PRMT1 binding partner, depletion clown-regulated arginine methylation in the nucleus and inhibited neurite outgrowth. These results indicate that protein arginine methylation by PRMT1 in the nucleus is an important step in neuritogenesis. (c) 2008 Elsevier Ireland Ltd. All rights reserved.
  • Alexander G. Bassuk; Robyn H. Wallace; Aimee Buhr; Andrew R. Buller; Zaid Afawi; Masahito Shimojo; Shingo Miyata; Shan Chen; Pedro Gonzalez-Alegre; Hilary L. Griesbach; Shu Wu; Marcus Nashelsky; Eszter K. Vladar; Dragana Antic; Polly J. Ferguson; Sebahattin Cirak; Thomas Voit; Matthew P. Scott; Jeffrey D. Axelrod; Christina Gurnett; Azhar S. Daoud; Sara Kivity; Miriam Y. Neufeld; Aziz Mazarib; Rachel Straussberg; Simri Walid; Amos D. Korczyn; Diane C. Slusarski; Samuel F. Berkovic; Hatem I. El-Shanti
    AMERICAN JOURNAL OF HUMAN GENETICS 83 5 572 - 581 2008年11月 [査読有り]
     
    Progressive myoclonus epilepsy (PME) is a syndrome characterized by myoclonic seizures (lightning-like jerks), generalized convulsive seizures, and varying degrees of neurological decline, especially ataxia and dementia. Previously, we characterized three pedigrees of individuals with PME and ataxia, where either clinical features or linkage mapping excluded known PME loci. This report identifies a mutation in PRICKLE1 (also known as RILP for REST/NRSF interacting LIM domain protein) in all three of these pedigrees. The identified PRICKLE1 mutation blocks the PRICKLE1 and REST interaction in vitro and disrupts the normal function of PRICKLE1 in an in vivo zebrafish overexpression system. PRICKLE1 is expressed in brain regions implicated in epilepsy and ataxia in mice and humans, and, to our knowledge, is the first molecule in the noncanonical WNT signaling pathway to be directly implicated in human epilepsy.
  • Hiroaki Okuda; Shingo Miyata; Yasutake Mori; Masaya Tohyama
    FEBS LETTERS 581 24 4754 - 4760 2007年10月 [査読有り]
     
    The Drosophila planar cell polarity (PCP) gene prickle has been previously indicated as one of the regulators of gastrulation in the early embryonic stage. However, the functional role of prickle in the brain in particular is not known. We first indicated that mouse Prickle1 and Prickle2 are continually expressed in the brain throughout the embryonic stages and are observed to be specifically expressed in the postmitotic neurons. Furthermore, Prickle1 or Prickle2 depletion effectively decreases the neurite outgrowth levels of mouse neuroblastoma Neuro2a cells. These results indicate that mouse Prickle1 and Prickle2 possibly regulate positive neurite formation during brain development. (c) 2007 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.
  • Tsuya Taneda; Shingo Miyata; Aoi Kousaka; Kiyoshi Inoue; Yoshihisa Koyama; Yasutake Mori; Masaya Tohyama
    BRAIN RESEARCH 1155 1 - 9 2007年06月 [査読有り]
     
    The regional distribution of PRMT8 transcript was examined in mouse brain using in situ hybridization (ISH) histochemistry. The PRMT8 cRNA probe was specifically hybridized with CNS and the signals were observed only in the neurons. The distribution of the neurons expressing PRMT8 mRNA was not even throughout the brain. All of the regions related to general somatosensory system expressed PRMT8 mRNA strongly. Most of the relay nuclei intervening the special somatosensory system, such as the auditory, visual, and vestibular systems, were packed with PRMT8 mRNA expressing neurons. Forebrain limbic areas and thalamic nuclei relevant to limbic areas were also strongly labeled with the probe. Some areas related to the motor system, such as the caudate putamen, Purkinje cells, inferior olivary nucleus and cerebellar nuclei expressed PRMT8 mRNA strongly. These findings suggest that PRMT8 is chiefly involved in the somatosensory and limbic systems, and a part of motor system.
  • T Fujiwara; Y Mori; DL Chu; Y Koyama; S Miyata; H Tanaka; K Yachi; T Kubo; H Yoshikawa; M Tohyama
    MOLECULAR AND CELLULAR BIOLOGY 26 6 2273 - 2285 2006年03月 [査読有り]
     
    HuD is an RNA-binding protein that has been shown to induce neuronal differentiation by stabilizing labile mRNAs carrying AU-rich instability elements. Here, we show a novel mechanism of arginine methylation of HuD by coactivator-associated arginine methyltransferase 1 (CARM1) that affected mRNA turnover of p21(cip1/waf1) mRNA in PC12 cells. CARM1 specifically methylated HuD in vitro and in vivo and colocalized with HuD in the cytoplasm. Inhibition of HuD methylation by CARM1 knockdown elongated the p21(cip1/waf1) mRNA half-life and resulted in a slow growth rate and robust neuritogenesis in response to nerve growth factor (NGF). Methylation-resistant HuD bound more p21(cip1/waf1) mRNA than did the wild type, and its overexpression upregulated p21(cip1/waf1) protein expression. These results suggested that CARM1-methylated HuD maintains PC12 cells in the proliferative state by committing p21(cip1/waf1) mRNA to its decay system. Since the methylated population of HuD was reduced in NGF-treated PC12 cells, downregulation of HuD methylation is a possible pathway through which NGF induces differentiation of PC12 cells.
  • K. Ikenaka; S. Miyata; Y. Mori; Y. Koyama; T. Taneda; H. Okuda; A. Kousaka; M. Tohyama
    NEUROSCIENCE 141 4 1971 - 1982 2006年 [査読有り]
     
    The distribution of protein arginine N-methyltransferase 3 (PRMT3) was investigated in the mouse brain using indirect immunofluorescence. PRMT3 was observed to be localized in the cell bodies and dendrites of neurons but not in the axons and glial cells, indicating that PRMT3 is involved in neuronal function. The distribution of the immunoreactive neurons in the brain was uneven, indicating that PRMT3 plays a role in specific neuronal systems such as the motor and limbic systems, as well as functions related to the cerebellum. The present ontogenetic analysis of PRMT1 and PRMT3 using Western blot methodology clearly revealed that PRMT3 develops during the perinatal stage and its expression is maintained even in adulthood. PRMT1, on the other hand, is expressed transiently during the early embryonic stage. These findings indicate that PRMT3 is related with neuronal function in both young and adult brains, while PRMT1 has roles in the immature brain, such as the formation of neural circuits. (c) 2006 IBRO. Published by Elsevier Ltd. All rights reserved.
  • S Miyata; Y Mori; T Fujiwara; K Ikenaka; S Matsuzaki; K Oono; T Katayama; M Tohyama
    MOLECULAR BRAIN RESEARCH 134 2 333 - 337 2005年04月 [査読有り]
     
    Localprotein synthesis in neuronal dendrites is one of the mechanisms that may mediate a rapid and synapse-specific mobilization of proteins from the resident mRNAs. A great deal of effort has been made in analyzing the dynamic state of protein synthesis in the living cells chiefly by quantifying protein level. However, the protein level cannot mirror the spatiotemporal alteration of translation because it can be affected, not only by protein synthesis, but also by other factors, like degradation. Therefore, it is problematic to visualize the dynamic state of translation by the present methods'. To solve the problem, we applied fluorescence resonance energy transfer (FRET) technique to in situ detection of the assembly and disassembly cycle among a pair of translation initiation factors [eukaryotic initiation factors (elFs)], thereby showing that BDNF and ephrin could potentiate local protein synthesis in the dendrites of hippocampal neurons. (c) 2004 Elsevier B.V All rights reserved.
  • K Oono; T Yoneda; T Manabe; S Yamagishi; S Matsuda; J Hitomi; S Miyata; T Mizuno; K Imaizumi; T Katayama; M Tohyama
    NEUROCHEMISTRY INTERNATIONAL 45 5 765 - 772 2004年10月 [査読有り]
     
    Recent papers have reported that neuronal death in patients with Alzheimer's disease, Parkinson's disease, and cerebral ischemia has its origin in the endoplasmic reticulum (ER). IRE lot is one of the ER stress transducers that detect the accumulation of unfolded proteins in the ER. IRE1alpha mediates two major cellular responses, which are the unfolded protein response (UPR), a defensive response, and apoptosis that leads to cell death. However, little is known about the regulatory mechanisms that select between the UPR and apoptosis. We identified Jun activation domain-binding protein-1 (JAB1) as a molecule that interacts with IRE1alpha using a yeast two-hybrid system. We demonstrated that JAB1 binds to IRE1alpha in the absence of stress, but that binding is decreased by ER stress inducers. Moreover, mutant JAB1 down-regulates the UPR signaling pathway through tight binding with IRE1alpha. These results suggested that JAB1 may act as a key molecule in selecting the UPR or cell death by association and dissociation with IRE I (X. (C) 2004 Elsevier Ltd. All rights reserved.
  • S Matsuzaki; T Manabe; T Katayama; A Nishikawa; T Yanagita; H Okuda; Y Yasuda; S Miyata; S Meshitsuka; M Tohyama
    JOURNAL OF NEUROCHEMISTRY 88 6 1345 - 1351 2004年03月 [査読有り]
     
    Oxidative stress is a major risk factor for Alzheimer's disease (AD) and other neurodegenerative disorders. Metals are known to be one of the factors that contribute to oxidative stress. Recently, we reported that the aberrant splicing isoform (PS2V) generated by skipping exon5 of the presenilin-2 (PS2) gene is a diagnostic feature of sporadic AD (SAD). PS2V is inducible by exposure of human neuroblastoma to hypoxia. We examined whether this aberrant splicing was caused by metal-induced oxidative stress, such as exposure to aluminum. As a result, we demonstrated that exposure to aluminum accelerated PS2V production induced by hypoxia. This acceleration of the production of PS2V to hypoxia was caused by chronic aluminum exposure, but was not related to the intracellular content of aluminum. HMGA1a is a mediator of PS2V production, and it was induced by aluminum as well as by hypoxia. Induction of HMGA1a was increased by chronic exposure to aluminum, and a nuclear extract containing HMGA1a bound to a specific sequence on exon5 of PS2 pre-mRNA, as reported previously. Finally, the acceleration of PS2V production induced by aluminum under hypoxic conditions reflected, but has not yet been directly shown to cause, vulnerability to endoplasmic reticulum stress. These results suggest that exposure to some metals can accelerate and enhance PS2V generation, and that hypoxia plus chronic exposure to metals may promote the development of AD.
  • T Katayama; K Imaizumi; T Yoneda; M Taniguchi; A Honda; T Manabe; J Hitomi; K Oono; K Baba; S Miyata; S Matsuzaki; K Takatsuji; M Tohyama
    CELLULAR AND MOLECULAR NEUROBIOLOGY 24 1 137 - 147 2004年02月 [査読有り]
     
    The human ADP-ribosylation factor-like protein, ARF4L is a member of the ARF family, which are small GTP-binding proteins that play significant roles in vesicle transport and protein secretion. However, little is known about the physiological roles of ARF4L. In this study, to understand the biological functions of ARF4L, we carried out immunocytochemical analysis of ARF4L molecules with mutations in the functional domains. ARF4L was shown to be distributed to the plasma membrane following binding to GTP (Q80L), and into endosomes following binding to GDP (T35N). Moreover, the inactive-form of ARF4L ( T35N) causes localization of transferrin receptors to the endosomal compartment, while the active form ( Q80L) causes transport to the plasma membrane. These findings indicate that ARF4L drive the transport of cargo protein and subsequent fusion of recycling vesicles with the plasma membrane for maintenance of the cell surface.
  • K Nemoto; S Miyata; F Nemoto; T Yasumoto; U Murai; H Kageyama; M Degawa
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 275 2 472 - 476 2000年08月 [査読有り]
     
    Neurotrophins including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3) are known to play important roles in the survival, proliferation, differentiation, and/or maintenance of function in several tissues including neuronal tissues. The role of neurotrophins in liver tissue, however, has not yet been clarified. In the present study, we assessed the temporal change in gene expression of neurotrophins, NGF, BDNF, and NT-3, and their receptors, low affinity neurotrophin receptor (p75NTR) and Trks A, B, and C, by RT-PCR technique in the liver of rats treated with lead nitrate (LN; 0.1 mmol/kg body weight), an inducer of liver hyperplasia. The mRNAs for NGF, BDNF with exon 4, NT-3, p75NTR, and all Trk members were detected in the LN-untreated liver. LNtreatment resulted in increases in the levels of NGF, BDNF with exon 4, NT-3, p75NTR, and TrkA mRNAs and further led to expression of BDNF mRNA with exon 3. The increase in gene expression of neurotrophins and their receptors was closely correlated with those in liver weight. In this report, we propose for the first time that neurotrophins may play crucial roles in LN-induced liver hyperplasia. (C) 2000 Academic Press.
  • K Nemoto; M Sekimoto; K Fukamachi; F Nemoto; S Miyata; Y Nakamura; M Hamada; E Senba; T Ueyama; M Degawa
    MOLECULAR BRAIN RESEARCH 68 1-2 186 - 189 1999年05月 [査読有り]
     
    We have previously reported that in cultured rat vascular smooth muscle cells (VSMCs), neurotrophin-3 (NT-3) gene expression was suppressed by TPA (12-O-tetradecanoyl phorbol-13-acetate), which induces an AP-1 transcription factor. In the present study, to clarify the mechanism for TPA-mediated downregulation of NT-3 gene expression, effects of cycloheximide and dexamethasone (Dex) on the TPA-mediated downregulation were examined in VSMCs. Pretreatment with cycloheximide, an inhibitor of protein synthesis, or simultaneous treatment with Dex, an inhibitor of AP-1, suppressed the TPA-mediated downregulation of NT-3 gene expression. Furthermore, co-transfection of c-fos and c-jun expression vectors into VSMCs resulted in decrease in the NT-3 gene expression. The present findings suggest that TPA-induced AP-1 de novo synthesis causes the downregulation of NT-3 gene expression in VSMCs. (C) 1999 Elsevier Science B.V. All rights reserved.
  • Sekimoto M; Fukamachi K; Nemoto F; Miyata S; Degawa M; Senba E; Ueyama T; Nemoto K
    NeuroReport 9 16 3675 - 3679 1998年11月 [査読有り]
     
    THE neurotrophin-3 (NT-3) gene has previously been reported to consist of three exons including two 5' short untranslated exons and a 3' long exon encoding the entire protein, and to give rise to two classes of transcripts by alternative splicing of the 5' exons to the 3' coding exon. In the present study, we demonstrated the presence of at least four new classes of transcripts of the NT-3 gene, in addition to the two known transcripts. The present finding proposes the further complexity of the regulational mechanism for NT-3 expression. NeuroReport 9: 3675-3679 (C) 1998 Lippincott Williams & Wilkins.
  • E Nemoto; E Fukamachi; F Nemoto; S Miyata; M Hamada; Y Nakamura; E Senba; T Ueyama
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 245 1 284 - 288 1998年04月 [査読有り]
     
    Most previous researches on neurotrophins including nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), and neurotrophin-3 (NT-3) have focused on the nervous system, because their receptors are widely distributed in neuronal tissues. Recently, however, the participation of neurotrophins in inflammation and atherosclerosis has been proposed. Therefore, the gene expression of neurotrophins is now an urgent issue is to be investigated in nonneuronal tissues. Here, we evaluated the gene expression of neurotrophins and their receptors in rat cultured vascular smooth muscle cells (VSMCs) by the reverse transcriptase-polymerase chain reaction method, The transcripts of NGF, NT-3, and TrkC thigh-affinity receptor for NT-3), and two BDNF alternative spliced transcript variants with exons 3 and 4 were clearly detected in VSMCs cultured under conventional culture conditions. The upregulation of mRNA levels for NGF, two BDNF variants with exons 1 and 2, low-affinity neurotrophin receptor, and high-affinity receptors, TrkA (for NGF) and TrkB (for BDNF), was observed in response to the treatment with serum and phorbol-ester following the serum-starvation. In contrast, the expression of NT-3 and TrkC genes was downregulated under these conditions. Go-expression of these factors and their receptors and the characteristic regulation of their gene transcriptions suggest that these factors play crucial roles in the function of VSMCs through an autocrine mechanism. (C) 1998 Academic Press.

MISC

書籍等出版物

  • ストレス科学事典
    宮田信吾; 遠山正彌 (担当:分担執筆範囲:)実務教育出版 2011年

講演・口頭発表等

  • 漢方薬の作用機序解明からみたトランスレーショナルリサーチ  [招待講演]
    宮田 信吾
    第76回日本自律神経学会総会 2023年10月 シンポジウム・ワークショップパネル(指名)
  • 更年期障害の精神神経症状を改善する加味逍遥散の作用機序解明  [通常講演]
    清水尚子; 石野雄吾; 小山佳久; 武田卓; 島田昌一; 遠山正彌; 宮田信吾
    第42回産婦人科漢方研究会 学術集会 2023年09月 口頭発表(一般)
  • アルギニンメチル化酵素 CARM1 によるオリゴデンドロサイト分化制御  [通常講演]
    石野 雄吾; 清水 尚子; 遠山 正彌; 宮田 信吾
    第21回ORIGIN神経科学懇話会 2023年08月 口頭発表(一般)
  • Regulations of oligodendrocyte differentiation and myelination by protein-arginine methyltransferases  [招待講演]
    石野 雄吾; 清水 尚子; 遠山 正彌; 宮田 信吾
    第64回日本神経病理学会総会学術研究会/第66回日本神経化学会大会 合同大会 2023年07月 シンポジウム・ワークショップパネル(公募)
  • Novel signal transduction mechanisms involved in oligodendrocyte differentiation and psychiatric disorders  [通常講演]
    宮田 信吾; 清水 尚子; 石野 雄吾; 遠山 正彌
    第64回日本神経病理学会総会学術研究会/第66回日本神経化学会大会 合同大会 2023年07月 シンポジウム・ワークショップパネル(公募)
  • 精神神経症状に対する抑肝散や加味逍遥散の作用機序へのmicroRNAsの関与  [招待講演]
    宮田信吾; 清水尚子; 石野雄吾; 遠山正彌
    第66回日本薬学会 関東支部大会 2022年09月 シンポジウム・ワークショップパネル(指名)
  • アルギニンメチル化酵素CARM1によるオリゴデンドロサイト分化制御
    石野雄吾; 清水尚子; 遠山正彌; 宮田信吾
    Neuro2022(第45回日本神経科学大会/第65回日本神経化学会大会/第32回日本神経回路学会大会) 2022年07月
  • 更年期障害における精神神経症状を改善する加味逍遥散の作用機序解明
    清水尚子; 石野雄吾; 武田卓; 遠山正彌; 宮田信吾
    第74回日本自律神経学会総会 2021年10月
  • 分子科学で精神疾患を解く  [招待講演]
    宮田信吾
    第40回 東邦大学 生命科学シンポジウム 「脳神経疾患への最新アプローチ」 2021年09月 公開講演,セミナー,チュートリアル,講習,講義等
  • 加味逍遥散の基礎的エビデンス  [招待講演]
    清水尚子; 石野雄吾; 武田卓; 遠山正彌; 宮田信吾
    第8回 Osaka Science-based Kampo研究会 2021年02月 公開講演,セミナー,チュートリアル,講習,講義等
  • オリゴデンドロサイト分化制御の新規分子機構解明へ向けて
    石野雄吾; 清水尚子; 遠山正彌; 宮田信吾
    第63回日本神経化学会大会 2020年09月 ポスター発表
  • うつ病のマウスモデルにおける人為的レム睡眠増加の影響
    安垣進之助; 柏木 光昭; 鹿糠 実香; 宮田 信吾; 柳沢正史; 林悠
    第43回日本神経科学大会 2020年08月 口頭発表(一般)
  • Yokukansan regulates microRNA expression and normalizes glucocorticoid receptor protein expression in the mice brain after stress exposure
    宮田信吾; 清水尚子; 石野雄吾; 遠山正彌
    第42回日本分子生物学会年会 2019年12月 ポスター発表
  • SSH「探求マナー講座」  [招待講演]
    宮田信吾
    日本分子生物学会 講師派遣事業 2019年11月 公開講演,セミナー,チュートリアル,講習,講義等
  • Discovery of novel antidepressant agents targeting protein kinases in oligodendrocytes
    S. Miyata; S. Shimizu; Y. Ishino; M. Tohyama
    Neuroscience 2019, SfN's 49th annual meeting, 2019年10月 ポスター発表
  • 抗ストレス効果に関連する低分子化合物の探索  [通常講演]
    宮田信吾; 清水尚子; 石野雄吾; 遠山正彌
    第20回ORIGIN神経科学研究会 2019年09月 口頭発表(一般)
  • 更年期障害における精神神経症状を改善する加味逍遥散の作用機序の解析  [通常講演]
    清水尚子; 石野雄吾; 遠山正彌; 宮田信吾
    第39回産婦人科漢方研究会学術集会 2019年09月 口頭発表(一般)
  • The relation between depression and REM sleep: insights from a model mouse of depression
    安垣進之助; 劉至堯; 柏木光昭; 鹿糠実香; 本多隆利; 宮田信吾; 柳沢正史; 林悠
    NEURO2019, 第42回日本神経科学大会、第62回日本神経化学大会 2019年07月 口頭発表(一般)
  • Towards understanding of novel molecular mechanisms underlying oligodendrocyte properties both in normal and pathological condition  [通常講演]
    Ishino Y; Shimizu S; Tohyama M; Miyata S
    Neuro2019, The 42nd Annual Meeting of the Japan Neuroscience Society, The 62nd Annual Meeting of the Japanese Society for Neurochemistry 2019年07月 シンポジウム・ワークショップパネル(公募)
  • 新たな創薬研究領域の開拓〜新技術から見えてきたオリゴデンドロサイトとシュワン細胞の共通性〜  [通常講演]
    宮田信吾; 服部剛志
    NEURO2019, 第42回日本神経科学大会、第62回日本神経化学大会 シンポジウム・オーガナイザー・座長 2019年07月 シンポジウム・ワークショップパネル(公募)
  • うつ病とレム睡眠の関係について―マウスのうつ病モデルを用いた解析―
    安垣進之助; 劉至堯; 柏木光昭; 鹿糠実香; 本多隆利; 宮田信吾; 柳沢正史; 林悠
    日本睡眠学会第44回定期学術集会 2019年06月 口頭発表(一般)
  • 脳内化学物質変化から捉える脳機能と疾患  [招待講演]
    宮田信吾
    近畿大学サイエンス・シンポジウム 躍動する物質科学・脳科学の最前線〜次世代の化学者への提言〜 2019年03月 シンポジウム・ワークショップパネル(指名)
  • 慢性ストレスにより誘発される脳白質異常の分子機構解析
    石野雄吾; 清水尚子; 遠山正彌; 宮田信吾
    第41回日本分子生物学会年会 2018年11月 ポスター発表
  • NDE1はオリゴデンドロサイトの分化を促進する
    清水尚子; 石野雄吾; 遠山正彌; 宮田信吾
    第41回日本分子生物学会年会 2018年11月 ポスター発表
  • うつ病発症における脳白質構造変化とオリゴデンドロサイト機能変化の関連  [通常講演]
    宮田信吾; 清水尚子; 石野雄吾; 山本彬世; 川上あゆみ; 遠山正彌
    2017年度生命科学系学会合同年次大会(ComBio2017) 2017年12月
  • 分子生物学を用いた最先端研究〜神経科学研究から創薬まで〜  [招待講演]
    宮田信吾
    日本分子生物学会 講師派遣事業 2017年12月 公開講演,セミナー,チュートリアル,講習,講義等
  • Molecular mechanisms of the onset of neuropsychiatric disorders by the changes in glial functions.  [通常講演]
    Shingo Miyata; Shoko Shimizu; Yugo Ishino; Masaya Tohyama
    The 60th Annual Meeting of the Japanese Society for Neurochemistry. 2017年09月
  • グリアと神経の成熟と精神・神経疾患  [通常講演]
    宮田信吾
    第60回日本神経化学会大会シンポジウム オーガナイザー・座長 2017年09月 シンポジウム・ワークショップパネル(指名)
  • 精神疾患関連分子によるオリゴデンドロサイト発達制御  [通常講演]
    宮田信吾; 清水尚子; 服部剛志; 遠山正彌
    第38回日本生物学的精神医学会・第59回日本神経化学会大会 合同年会 2016年09月
  • 脳白質機能変化とうつ病発症の関連性〜マウスモデル動物からヒトうつ病患者まで〜  [招待講演]
    宮田信吾
    神戸大学大学院医学系研究科 先端医学トピックス 2016年07月
  • 漢方薬はなぜ効くか?〜基礎医学から〜  [招待講演]
    宮田信吾
    第22回日本女性心身医学会研修会・講演 2016年02月
  • うつ病発症に関わるオリゴデンドロサイトの機能異常に関する研究  [通常講演]
    宮田信吾; 清水尚子; 田中貴士; 松村彬世; 川上あゆみ; 鹿島美恵子; 遠山正彌
    BMB2015, 第38回日本分子生物学会年会、第88回日本生化学会大会 合同大会 2015年12月
  • Plasma corticosterone regulates cadherin superfamily molecules in oligodendrocytes in the mouse corpus callosum  [通常講演]
    Shingo Miyata; Keiko Yoshikawa; Manabu Taniguchi; Toshiko Ishikawa; Takashi Tanaka; Shoko Shimizu; Masaya Tohyama
    第20回グリア研究会 2015年12月
  • 慢性ストレス応答と抑肝散による自律神経系制御の可能性  [通常講演]
    宮田信吾; 清水尚子; 田中貴士; 松村彬世; 川上あゆみ; 遠山正彌
    第68回日本自律神経学会総会 2015年10月
  • 基礎医学研究における漢方薬のエビデンス~抑肝散の精神神経症状に対する効果の検討~  [招待講演]
    宮田信吾
    第14回愛媛東洋医学カンファレンス(COME) 2015年02月
  • ゴルジストレス特異的発現変動因子の機能解析  [通常講演]
    宮田信吾; 遠山正彌
    第37回日本分子生物学会年会 2014年12月
  • ストレス応答系に対する抑肝散の効果  [通常講演]
    宮田信吾
    第41回日本脳科学会 2014年11月
  • オリゴデンドロサイトの機能とストレス  [通常講演]
    宮田信吾; 清水尚子; 田中貴士; 遠山正彌
    第36回日本生物学的精神医学会、第57回日本神経化学会大会 合同学会 2014年09月
  • エビデンスに基づいた漢方薬の基礎医学研究~神経系への抑肝散の効果~  [招待講演]
    宮田信吾
    第2回 Osaka Science-based Kampo研究会 2014年02月 公開講演,セミナー,チュートリアル,講習,講義等
  • 慢性ストレスによる線維束オリゴデンドロサイトの構造変化  [通常講演]
    宮田信吾; 清水尚子; 遠山正彌
    第18回グリア研究会 2013年10月 口頭発表(一般) 仙台 グリア研究会
  • うつ病発症に関わる脳白質特異的シグナル伝達経路の解明  [通常講演]
    宮田信吾; 清水尚子; 遠山正彌
    第72回近畿大学医学会学術講演会 2013年 大阪 近畿大学医学会
  • 漢方薬と神経細胞死 -抑肝散の神経機能障害への効能解析-  [通常講演]
    宮田信吾
    第34回日本生物学的精神医学会 2012年09月
  • こころの病気・脳の病気の科学的解析  [通常講演]
    宮田信吾
    近畿大学医学部 基礎医学系セミナー 2012年07月 公開講演,セミナー,チュートリアル,講習,講義等 大阪 近畿大学医学部
  • うつ病態におけるグリア細胞の役割  [招待講演]
    宮田信吾
    CRESTセミナー 2012年04月 公開講演,セミナー,チュートリアル,講習,講義等 奈良 奈良先端科学技術大学
  • 慢性ストレス応答によるオリゴデンドロサイト特異的なシグナル伝達機構の解明  [通常講演]
    宮田信吾; 小山佳久; 吉川景子; 石川淑子; 谷口学; 嶽本香菜; 遠山正彌
    第15回 グリア研究会 2010年10月 福岡 グリア研究会
  • Molecular pathogenesis of major depression: The relationship between oligodendrocyte dysfunctions  [通常講演]
    Shingo Miyata
    Neuro2010 第33回日本神経科学大会、第53回日本神経化学会大会、第20回日本神経回路学会大会合同大会 2010年09月 シンポジウム・ワークショップパネル(公募) 神戸 日本神経化学会
  • オリゴデンドロサイト特異的な発現変動因子によるうつ病発症機構の解明  [通常講演]
    宮田信吾; 吉川景子; 嶽本香奈; 小山佳久; 遠山正彌
    第14回 グリア研究会 2009年11月 大阪 グリア研究会
  • 神経細胞におけるミトコンドリア局在TRAP1とERストレスとの関連性  [通常講演]
    嶽本香奈; 宮田信吾; 吉川景子; 片山泰一; 遠山正彌
    第85回 日本解剖学会近畿支部学術集会 2009年11月 奈良 日本解剖学会
  • 精神疾患発症機構へのmicroRNAの関与  [通常講演]
    宮田信吾; 橋本亮太; 嶽本香菜; 吉川景子; 遠山正彌
    第62回 日本自律神経学会総会 2009年11月 和歌山 自律神経学会
  • alpha CaMKII mRNAの輸送機構に関与するタンパク質の同定  [通常講演]
    森泰丈; 宮田信吾; 遠山正彌
    第52回 日本神経化学会大会 2009年06月
  • ラット海馬神経細胞での局所的翻訳におけるrpS2の安定化の重要性  [通常講演]
    宮田信吾; 森泰丈; 遠山正彌
    第52回 日本神経化学会大会 2009年06月 群馬 日本神経化学会
  • ヒト脳におけるserum/glucocorticoid regulated kinase 1 (Sgk1) との相互作用因子の網羅的探索  [通常講演]
    石川淑子; 宮田信吾; 吉川景子; 嶽本香菜; 遠山正彌
    第115回 日本解剖学会総会 2009年03月
  • 慢性ストレス応答によるオリゴデンドロサイト機能異常の可能性  [通常講演]
    宮田信吾; 吉川景子; 石川淑子; 嶽本香奈; 谷口学; 小山佳久; 片山泰一; 遠山正彌
    第115回 日本解剖学会総会 2009年03月 岩手 日本解剖学会
  • タンパクメチル化酵素Protein Arginine N-methyltransferase 3 (PRMT3) によるラット海馬神経細胞樹状突起での翻訳制御機構  [通常講演]
    宮田信吾; 森泰丈; 遠山正彌
    第51回 日本神経化学会大会 2008年09月
  • 神経細胞の形態分化におけるタンパク質アルギニンメチル化の機能解析—タンパク質アルギニンメチル化酵素 (PRMT) ノックダウン細胞の解析から分かったこと  [招待講演]
    宮田信吾
    第78回 千里神経懇話会・新適塾 2006年10月 公開講演,セミナー,チュートリアル,講習,講義等
  • 神経分化におけるタンパクメチル化の関与の可能性  [通常講演]
    宮田信吾; 森泰丈; 藤原達司; 遠山正彌
    第26回 日本分子生物学会年会 2003年12月

担当経験のある科目_授業

  • 医学概論 演習シリーズ近畿大学
  • 医学部チュートリアル演習(機能I)近畿大学
  • 医学部プロフェッショナリズム講義:基礎配属実習近畿大学
  • 解剖生理学看護専門学校
  • 生理学I大阪大学
  • 情報活用基礎大阪大学
  • 発生学大阪大学
  • 神経科学特論大阪大学
  • 組織学・組織学実習大阪大学、近畿大学
  • 神経解剖学・脳実習大阪大学
  • 系統解剖学・解剖学実習大阪大学、近畿大学

所属学協会

  • 日本抗加齢学会   日本RNA学会   日本自律神経学会   日本分子生物学会   日本生化学会   日本解剖学会   日本神経科学学会   日本神経化学会   米国神経科学会   

共同研究・競争的資金等の研究課題

  • 日本学術振興会:科学研究費助成事業
    研究期間 : 2023年04月 -2026年03月 
    代表者 : 宮田 信吾; 遠山 正彌; 清水 尚子; 石野 雄吾
  • コロナ禍における孤独とストレスへの近大発対処法の創出
    近畿大学:”オール近大”新型コロナウイルス感染症対策支援プロジェクト
    研究期間 : 2021年08月 -2022年03月 
    代表者 : 清水尚子; 宮田信吾; 石野雄吾; 遠山正彌
  • 新規うつ病治療薬の創出
    大阪大学医学部附属病院未来医療開発部:ACT Japan 橋渡し研究戦略的推進プログラム
    研究期間 : 2020年04月 -2022年03月 
    代表者 : 宮田信吾、清水尚子、石野雄吾、遠山正彌
  • ストレス性精神症状改善に向けた抑肝散によるmicroRNA制御機構の解明
    文部科学省:基盤研究(C)
    研究期間 : 2019年04月 -2022年03月 
    代表者 : 遠山正彌; 宮田信吾; 清水尚子; 石野雄吾
  • Withコロナ生活へ向けて〜自粛生活による孤独感と運動不足がもたらす精神神経化学的影響の分子機序解明およびAll近大goodsによる改善効果の検討
    近畿大学:”オール近大”新型コロナウイルス感染症対策支援プロジェクト
    研究期間 : 2020年08月 -2021年03月 
    代表者 : 宮田信吾; 清水尚子; 石野雄吾; 遠山正彌
  • 腸内細菌叢・脳軸からみた月経前症候群への新治療戦略
    文部科学省:基盤研究(C)
    研究期間 : 2019年04月 -2021年03月 
    代表者 : 武田卓; 清水尚子; 平野昌美; 宮田信吾; 石野雄吾
  • 慢性ストレス応答に関連する脳内PI3Kシグナルの重要性
    公益財団法人 日本応用酵素協会:酵素研究助成
    研究期間 : 2018年04月 -2019年03月 
    代表者 : 宮田信吾
  • オリゴデンドロサイト機能異常よるうつ病発症機構へのタンパクメチル化の関与
    文部科学省:基盤研究(C)
    研究期間 : 2016年04月 -2019年03月 
    代表者 : 宮田信吾
  • 細胞増殖や浸潤シグナル経路を抑制する生薬成分の探索・同定
    国立大学法人 富山大学和漢医薬学総合研究所:共同利用・共同研究:探索研究プロジェクト
    研究期間 : 2017年04月 -2018年03月 
    代表者 : 宮田信吾
  • アストロサイトに高発現するDISC1の機能解析~統合失調症との関連を中心に~
    文部科学省:基盤研究(C)
    研究期間 : 2015年04月 -2018年03月 
    代表者 : 遠山正彌
  • 新規うつ病治療薬の探索
    日本医療研究開発機構:創薬総合支援事業
    研究期間 : 2014年11月 -2018年03月 
    代表者 : 宮田信吾
  • PMS症状を標的とした女性アスリートパフォーマンス改善への栄養学的アプローチ
    文部科学省:基盤研究(C)
    研究期間 : 2015年04月 -2017年03月 
    代表者 : 武田卓
  • 髄鞘による軸索機能制御に関わる細胞内・細胞間情報伝達機構の解明
    文部科学省:新学術領域研究(研究領域提案型)
    研究期間 : 2014年04月 -2016年03月 
    代表者 : 宮田信吾
  • 脳白質におけるリン酸化とメチル化をキーとしたうつ病発症機構の解明
    文部科学省:基盤研究(C)
    研究期間 : 2013年04月 -2016年03月 
    代表者 : 宮田信吾
  • 文部科学省:若手研究(B)
    研究期間 : 2008年04月 -2011年03月 
    代表者 : 宮田信吾
  • 抑肝散の示す精神疾患周辺行動改善に対する科学的検証
    厚生労働省:厚生労働科学研究費補助金(厚生科研費)
    研究期間 : 2008年04月 -2010年03月 
    代表者 : 遠山正彌
  • 神経細胞における樹状突起へのmRNA輸送機構とタンパクメチル化に関する研究
    文部科学省:特別研究員奨励費
    研究期間 : 2002年04月 -2005年03月 
    代表者 : 宮田信吾

その他のリンク