Updated on 2024/02/29

写真a

 
TAKASUGI Nobumasa
 
Organization
Faculty of Medicine, Dentistry and Pharmaceutical Sciences Associate Professor
Position
Associate Professor
External link

Degree

  • 博士(薬学) ( 2003.3   東京大学 )

  • Ph.D. ( The University of Tokyo )

Research Interests

  • Alzheimer disease

  • Neurodegenerative disease

  • vesicular transport

  • Amyloid beta

  • lipid signaling

Research Areas

  • Life Science / Pharmaceutical hygiene and biochemistry

  • Life Science / Pharmacology

Education

  • 東京大学大学院   薬学系研究科   薬学科

    1999.4 - 2003.3

      More details

Research History

  • Okayama University   医歯薬学系総合研究科   Associate Professor

    2015.7

      More details

  • Juntendo University   医学系研究科   Assistant Professor

    2012.4 - 2015.6

      More details

  • シカゴ大学   博士研究員

    2010.10 - 2012.3

      More details

  • 東京大学大学院   助教

    2010.4 - 2010.9

      More details

  • 東京大学大学院   助教

    2008.4 - 2010.3

      More details

  • 東京大学大学院   研究員

    2006.4 - 2008.3

      More details

▼display all

Professional Memberships

Committee Memberships

  • 日本薬学会薬理系薬学部会   会計実務  

    2023.4   

      More details

  • 日本薬理学会   学術評議員  

    2019   

      More details

  • 日本NO学会   庶務幹事  

    2018 - 2022   

      More details

 

Papers

  • Nuclear SphK2/S1P signaling is a key regulator of ApoE production and Aβ uptake in astrocytes Reviewed

    Masato Komai, Yuka Noda, Atsuya Ikeda, Nanaka Kaneshiro, Yuji Kamikubo, Takashi Sakurai, Takashi Uehara, Nobumasa Takasugi

    Journal of Lipid Research   2024.3

     More details

    Authorship:Last author, Corresponding author   Publishing type:Research paper (scientific journal)  

    DOI: 10.1016/j.jlr.2024.100510

    researchmap

  • Methyl vinyl ketone and its analogs covalently modify PI3K and alter physiological functions by inhibiting PI3K signaling Reviewed

    Atsushi Morimoto, Nobumasa Takasugi, Yuexuan Pan, Sho Kubota, Naoshi Dohmae, Yumi Abiko, Koji Uchida, Yoshito Kumagai, Takashi Uehara

    Journal of Biological Chemistry   2024.3

     More details

    Publishing type:Research paper (scientific journal)  

    DOI: 10.1016/j.jbc.2024.105679

    researchmap

  • Characterization of pathological changes in the olfactory system of mice exposed to methylmercury Reviewed

    Yuta Iijima, Ryohei Miki, Nobumasa Takasugi, Masatake Fujimura, Takashi Uehara

    Archives of Toxicology   2024.2

     More details

    Publishing type:Research paper (scientific journal)   Publisher:Springer Science and Business Media LLC  

    Abstract

    Methylmercury (MeHg) is a well-known environmental neurotoxicant that causes severe brain disorders such as Minamata disease. Although some patients with Minamata disease develop olfactory dysfunction, the underlying pathomechanism is largely unknown. We examined the effects of MeHg on the olfactory system using a model of MeHg poisoning in which mice were administered 30 ppm MeHg in drinking water for 8 weeks. Mice exposed to MeHg displayed significant mercury accumulation in the olfactory pathway, including the nasal mucosa, olfactory bulb, and olfactory cortex. The olfactory epithelium was partially atrophied, and olfactory sensory neurons were diminished. The olfactory bulb exhibited an increase in apoptotic cells, hypertrophic astrocytes, and amoeboid microglia, mainly in the granular cell layer. Neuronal cell death was observed in the olfactory cortex, particularly in the ventral tenia tecta. Neuronal cell death was also remarkable in higher-order areas such as the orbitofrontal cortex. Correlation analysis showed that neuronal loss in the olfactory cortex was strongly correlated with the plasma mercury concentration. Our results indicate that MeHg is an olfactory toxicant that damages the central regions involved in odor perception. The model described herein is useful for analyzing the mechanisms and treatments of olfactory dysfunction in MeHg-intoxicated patients.

    DOI: 10.1007/s00204-024-03682-w

    researchmap

    Other Link: https://link.springer.com/article/10.1007/s00204-024-03682-w/fulltext.html

  • Attenuation of protein arginine dimethylation via S-nitrosylation of protein arginine methyltransferase 1 Reviewed

    Rikako Taniguchi, Yuto Moriya, Naoshi Dohmae, Takehiro Suzuki, Kengo Nakahara, Sho Kubota, Nobumasa Takasugi, Takashi Uehara

    Journal of Pharmacological Sciences   2023.12

     More details

    Publishing type:Research paper (scientific journal)   Publisher:Elsevier BV  

    DOI: 10.1016/j.jphs.2023.12.012

    researchmap

  • Pivotal role for S-nitrosylation of DNA methyltransferase 3B in epigenetic regulation of tumorigenesis Reviewed International journal

    Kosaku Okuda, Kengo Nakahara, Akihiro Ito, Yuta Iijima, Ryosuke Nomura, Ashutosh Kumar, Kana Fujikawa, Kazuya Adachi, Yuki Shimada, Satoshi Fujio, Reina Yamamoto, NOBUMASA TAKASUGI, Kunishige Onuma, Mitsuhiko Osaki, Futoshi Okada, Taichi Ukegawa, Yasuo Takeuchi, Norihisa Yasui, Atsuko Yamashita, Hiroyuki Marusawa, Yosuke Matsushita, Toyomasa Katagiri, Takahiro Shibata, Koji Uchida, Sheng-Yong Niu, Nhi Lang, Tomohiro Nakamura, Kam Zhang, Stuart Lipton, Takashi Uehara

    Nature Communications   14 ( 1 )   621 - 621   2023.2

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:Springer Science and Business Media {LLC}  

    <jats:title>Abstract</jats:title><jats:p>DNA methyltransferases (DNMTs) catalyze methylation at the C5 position of cytosine with <jats:italic>S</jats:italic>-adenosyl-<jats:sc>l</jats:sc>-methionine. Methylation regulates gene expression, serving a variety of physiological and pathophysiological roles. The chemical mechanisms regulating DNMT enzymatic activity, however, are not fully elucidated. Here, we show that protein S-nitrosylation of a cysteine residue in DNMT3B attenuates DNMT3B enzymatic activity and consequent aberrant upregulation of gene expression. These genes include Cyclin D2 (<jats:italic>Ccnd2</jats:italic>), which is required for neoplastic cell proliferation in some tumor types. In cell-based and in vivo cancer models, only DNMT3B enzymatic activity, and not DNMT1 or DNMT3A, affects <jats:italic>Ccnd2</jats:italic> expression. Using structure-based virtual screening, we discovered chemical compounds that specifically inhibit <jats:italic>S</jats:italic>-nitrosylation without directly affecting DNMT3B enzymatic activity. The lead compound, designated DBIC, inhibits <jats:italic>S</jats:italic>-nitrosylation of DNMT3B at low concentrations (IC<jats:sub>50</jats:sub> ≤ 100 nM). Treatment with DBIC prevents nitric oxide (NO)-induced conversion of human colonic adenoma to adenocarcinoma in vitro. Additionally, in vivo treatment with DBIC strongly attenuates tumor development in a mouse model of carcinogenesis triggered by inflammation-induced generation of NO. Our results demonstrate that de novo DNA methylation mediated by DNMT3B is regulated by NO, and DBIC protects against tumor formation by preventing aberrant <jats:italic>S</jats:italic>-nitrosylation of DNMT3B.</jats:p>

    DOI: 10.1038/s41467-023-36232-6

    PubMed

    researchmap

  • The Pursuit of the “Inside” of the Amyloid Hypothesis—Is C99 a Promising Therapeutic Target for Alzheimer’s Disease? Invited Reviewed

    Nobumasa Takasugi, Masato Komai, Nanaka Kaneshiro, Atsuya Ikeda, Yuji Kamikubo, Takashi Uehara

    Cells   12 ( 3 )   454 - 454   2023.1

     More details

    Authorship:Lead author, Corresponding author   Publishing type:Research paper (scientific journal)   Publisher:MDPI AG  

    Aducanumab, co-developed by Eisai (Japan) and Biogen (U.S.), has received Food and Drug Administration approval for treating Alzheimer’s disease (AD). In addition, its successor antibody, lecanemab, has been approved. These antibodies target the aggregated form of the small peptide, amyloid-β (Aβ), which accumulates in the patient brain. The “amyloid hypothesis” based therapy that places the aggregation and toxicity of Aβ at the center of the etiology is about to be realized. However, the effects of immunotherapy are still limited, suggesting the need to reconsider this hypothesis. Aβ is produced from a type-I transmembrane protein, Aβ precursor protein (APP). One of the APP metabolites, the 99-amino acids C-terminal fragment (C99, also called βCTF), is a direct precursor of Aβ and accumulates in the AD patient’s brain to demonstrate toxicity independent of Aβ. Conventional drug discovery strategies have focused on Aβ toxicity on the “outside” of the neuron, but C99 accumulation might explain the toxicity on the “inside” of the neuron, which was overlooked in the hypothesis. Furthermore, the common region of C99 and Aβ is a promising target for multifunctional AD drugs. This review aimed to outline the nature, metabolism, and impact of C99 on AD pathogenesis and discuss whether it could be a therapeutic target complementing the amyloid hypothesis.

    DOI: 10.3390/cells12030454

    researchmap

  • Ex vivo analysis platforms for monitoring amyloid precursor protein cleavage Reviewed

    Yuji Kamikubo, Hao Jin, Yiyao Zhou, Kazue Niisato, Yoshie Hashimoto, Nobumasa Takasugi, Takashi Sakurai

    Frontiers in Molecular Neuroscience   15   2023.1

     More details

    Publishing type:Research paper (scientific journal)   Publisher:Frontiers Media SA  

    Alzheimer’s disease (AD) is a progressive neurodegenerative brain disorder and the most common cause of dementia in the elderly. The presence of large numbers of senile plaques, neurofibrillary tangles, and cerebral atrophy is the characteristic feature of AD. Amyloid β peptide (Aβ), derived from the amyloid precursor protein (APP), is the main component of senile plaques. AD has been extensively studied using methods involving cell lines, primary cultures of neural cells, and animal models; however, discrepancies have been observed between these methods. Dissociated cultures lose the brain’s tissue architecture, including neural circuits, glial cells, and extracellular matrix. Experiments with animal models are lengthy and require laborious monitoring of multiple parameters. Therefore, it is necessary to combine these experimental models to understand the pathology of AD. An experimental platform amenable to continuous observation and experimental manipulation is required to analyze long-term neuronal development, plasticity, and progressive neurodegenerative diseases. In the current study, we provide a practical method to slice and cultivate rodent hippocampus to investigate the cleavage of APP and secretion of Aβ in an ex vivo model. Furthermore, we provide basic information on Aβ secretion using slice cultures. Using our optimized method, dozens to hundreds of long-term stable slice cultures can be coordinated simultaneously. Our findings are valuable for analyses of AD mouse models and senile plaque formation culture models.

    DOI: 10.3389/fnmol.2022.1068990

    researchmap

  • Alterations in UPR Signaling by Methylmercury Trigger Neuronal Cell Death in the Mouse Brain Reviewed

    Ryosuke Nomura, Nobumasa Takasugi, Hideki Hiraoka, Yuta Iijima, Takao Iwawaki, Yoshito Kumagai, Masatake Fujimura, Takashi Uehara

    International Journal of Molecular Sciences   23 ( 23 )   15412 - 15412   2022.12

     More details

    Publishing type:Research paper (scientific journal)   Publisher:MDPI AG  

    Methylmercury (MeHg), an environmental toxicant, induces neuronal cell death and injures specific areas of the brain. MeHg is known to induce oxidative and endoplasmic reticulum (ER) stress. The unfolded protein response (UPR) pathway has a dual nature in that it regulates and protects cells from an overload of improperly folded proteins in the ER, whereas excessively stressed cells are eliminated by apoptosis. Oxidative stress/ER stress induced by methylmercury exposure may tilt the UPR toward apoptosis, but there is little in vivo evidence of a direct link to actual neuronal cell death. Here, by using the ER stress-activated indicator (ERAI) system, we investigated the time course signaling alterations of UPR in vivo in the most affected areas, the somatosensory cortex and striatum. In the ERAI-Venus transgenic mice exposed to MeHg (30 or 50 ppm in drinking water), the ERAI signal, which indicates the activation of the cytoprotective pathway of the UPR, was only transiently enhanced, whereas the apoptotic pathway of the UPR was persistently enhanced. Furthermore, detailed analysis following the time course showed that MeHg-induced apoptosis is strongly associated with alterations in UPR signaling. Our results suggest that UPR modulation could be a therapeutic target for treating neuropathy.

    DOI: 10.3390/ijms232315412

    researchmap

  • Lipid flippase dysfunction as a therapeutic target for endosomal anomalies in Alzheimer's disease. Reviewed International journal

    Nanaka Kaneshiro, Masato Komai, Ryosuke Imaoka, Atsuya Ikeda, Yuji Kamikubo, Takashi Saito, Takaomi C Saido, Taisuke Tomita, Tadafumi Hashimoto, Takeshi Iwatsubo, Takashi Sakurai, Takashi Uehara, Nobumasa Takasugi

    iScience   25 ( 3 )   103869 - 103869   2022.3

     More details

    Authorship:Last author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)  

    Endosomal anomalies because of vesicular traffic impairment have been indicated as an early pathology of Alzheimer'| disease (AD). However, the mechanisms and therapeutic targets remain unclear. We previously reported that βCTF, one of the pathogenic metabolites of APP, interacts with TMEM30A. TMEM30A constitutes a lipid flippase with P4-ATPase and regulates vesicular trafficking through the asymmetric distribution of phospholipids. Therefore, the alteration of lipid flippase activity in AD pathology has got attention. Herein, we showed that the interaction between βCTF and TMEM30A suppresses the physiological formation and activity of lipid flippase in AD model cells, A7, and AppNL-G-F/NL-G-F model mice. Furthermore, the T-RAP peptide derived from the βCTF binding site of TMEM30A improved endosomal anomalies, which could be a result of the restored lipid flippase activity. Our results provide insights into the mechanisms of vesicular traffic impairment and suggest a therapeutic target for AD.

    DOI: 10.1016/j.isci.2022.103869

    PubMed

    researchmap

  • Spatiotemporal analysis of the UPR transition induced by methylmercury in the mouse brain. Reviewed International journal

    Hideki Hiraoka, Ryosuke Nomura, Nobumasa Takasugi, Ryoko Akai, Takao Iwawaki, Yoshito Kumagai, Masatake Fujimura, Takashi Uehara

    Archives of toxicology   95 ( 4 )   1241 - 1250   2021.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Methylmercury (MeHg), an environmental toxicant, induces neuronal cell death and injures a specific area of the brain. MeHg-mediated neurotoxicity is believed to be caused by oxidative stress and endoplasmic reticulum (ER) stress but the mechanism by which those stresses lead to neuronal loss is unclear. Here, by utilizing the ER stress-activated indicator (ERAI) system, we investigated the signaling alterations in the unfolded protein response (UPR) prior to neuronal apoptosis in the mouse brain. In ERAI transgenic mice exposed to MeHg (25 mg/kg, S.C.), the ERAI signal, which indicates activation of the cytoprotective pathway of the UPR, was detected in the brain. Interestingly, detailed ex vivo analysis showed that the ERAI signal was localized predominantly in neurons. Time course analysis of MeHg exposure (30 ppm in drinking water) showed that whereas the ERAI signal was gradually attenuated at the late phase after increasing at the early phase, activation of the apoptotic pathway of the UPR was enhanced in proportion to the exposure time. These results suggest that MeHg induces not only ER stress but also neuronal cell death via a UPR shift. UPR modulation could be a therapeutic target for treating neuropathy caused by electrophiles similar to MeHg.

    DOI: 10.1007/s00204-021-02982-9

    PubMed

    researchmap

  • Covalent N-arylation by the pollutant 1,2-naphthoquinone activates the EGF receptor Reviewed

    Kengo Nakahara, Kyohei Hamada, Tomoki Tsuchida, Nobumasa Takasugi, Yumi Abiko, Kazuhiko Shien, Shinichi Toyooka, Yoshito Kumagai, Takashi Uehara

    Journal of Biological Chemistry   296   2021.1

     More details

    Publishing type:Research paper (scientific journal)  

    The epidermal growth factor receptor (EGFR) is the most intensively investigated receptor tyrosine kinase. Several EGFR mutations and modifications have been shown to lead to abnormal self-activation, which plays a critical role in carcinogenesis. Environmental air pollutants, which are associated with cancer and respiratory diseases, can also activate EGFR. Specifically, the environmental electrophile 1,2- naphthoquinone (1,2-NQ), a component of diesel exhaust particles and particulate matter more generally, has previously been shown to impact EGFR signaling. However, the detailed mechanism of 1,2-NQ function is unknown. Here, we demonstrate that 1,2-NQ is a novel chemical activator of EGFR but not other EGFR family proteins. We found that 1,2-NQ forms a covalent bond, in a reaction referred to as N-arylation, with Lys80, which is in the ligand-binding domain. This modification activates the EGFR-Akt signaling pathway, which inhibits serum deprivation-induced cell death in a human lung adenocarcinoma cell line. Our study reveals a novel mode of EGFR pathway activation and suggests a link between abnormal EGFR activation and environmental pollutant- associated diseases such as cancer.

    DOI: 10.1016/j.jbc.2021.100524

    Scopus

    PubMed

    researchmap

  • S-Nitrosylation at the active site decreases the ubiquitin-conjugating activity of ubiquitin-conjugating enzyme E2 D1 (UBE2D1), an ERAD-associated protein. Reviewed International journal

    Kana Fujikawa, Kengo Nakahara, Nobumasa Takasugi, Tadashi Nishiya, Akihiro Ito, Koji Uchida, Takashi Uehara

    Biochemical and biophysical research communications   524 ( 4 )   910 - 915   2020.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    S-Nitrosylation of protein cysteine thiol is a post-translational modification mediated by nitric oxide (NO). The overproduction of NO causes nitrosative stress, which is known to induce endoplasmic reticulum (ER) stress. We previously reported that S-nitrosylation of protein disulfide isomerase (PDI) and the ER stress sensor inositol-requiring enzyme 1 (IRE1) decreases their enzymatic activities. However, it remains unclear whether nitrosative stress affects ER-associated degradation (ERAD), a separate ER stress regulatory system responsible for the degradation of substrates via the ubiquitin-proteasomal pathway. In the present study, we found that the ubiquitination of a known ERAD substrate, serine/threonine-protein kinase 1 (SGK1), is attenuated by nitrosative stress. C-terminus of Hsc70-interacting protein (CHIP) together with ubiquitin-conjugating enzyme E2 D1 (UBE2D1) are involved in this modification. We detected that UBE2D1 is S-nitrosylated at its active site, Cys85 by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Furthermore, in vitro and cell-based experiments revealed that S-nitrosylated UBE2D1 has decreased ubiquitin-conjugating activity. Our results suggested that nitrosative stress interferes with ERAD, leading to prolongation of ER stress by co-disruption of various pathways, including the molecular chaperone and ER stress sensor pathways. Given that nitrosative stress and ER stress are upregulated in the brains of patient with Parkinson's disease (PD) and of those with Alzheimer's disease (AD), our findings may provide further insights into the pathogenesis of these neurodegenerative disorders.

    DOI: 10.1016/j.bbrc.2020.02.011

    PubMed

    researchmap

  • The Emerging Role of Electrophiles as a Key Regulator for Endoplasmic Reticulum (ER) Stress. Reviewed International journal

    Nobumasa Takasugi, Hideki Hiraoka, Kengo Nakahara, Shiori Akiyama, Kana Fujikawa, Ryosuke Nomura, Moeka Furuichi, Takashi Uehara

    International journal of molecular sciences   20 ( 7 )   2019.4

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)  

    The unfolded protein response (UPR) is activated by the accumulation of misfolded proteins in the endoplasmic reticulum (ER), which is called ER stress. ER stress sensors PERK, IRE1, and ATF6 play a central role in the initiation and regulation of the UPR; they inhibit novel protein synthesis and upregulate ER chaperones, such as protein disulfide isomerase, to remove unfolded proteins. However, when recovery from ER stress is difficult, the UPR pathway is activated to eliminate unhealthy cells. This signaling transition is the key event of many human diseases. However, the precise mechanisms are largely unknown. Intriguingly, reactive electrophilic species (RES), which exist in the environment or are produced through cellular metabolism, have been identified as a key player of this transition. In this review, we focused on the function of representative RES: nitric oxide (NO) as a gaseous RES, 4-hydroxynonenal (HNE) as a lipid RES, and methylmercury (MeHg) as an environmental organic compound RES, to outline the relationship between ER stress and RES. Modulation by RES might be a target for the development of next-generation therapy for ER stress-associated diseases.

    DOI: 10.3390/ijms20071783

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0002-6059-2082

  • Attenuation of Macrophage Migration Inhibitory Factor-Stimulated Signaling via S-Nitrosylation. Reviewed

    Kengo Nakahara, Kana Fujikawa, Hideki Hiraoka, Ikuko Miyazaki, Masato Asanuma, Akihiro Ito, Nobumasa Takasugi, Takashi Uehara

    Biological & pharmaceutical bulletin   42 ( 6 )   1044 - 1047   2019

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Nitric oxide (NO) is a key signaling molecule that has various effects via S-nitrosylation, a reversible post-translational modification that affects the enzymatic activity, localization, and metabolism of target proteins. As chronic nitrosative stress correlates with neurodegeneration, the targets have received focused attention. Macrophage migration inhibitory factor (MIF) plays a pivotal role in the induction of gene expression to control inflammatory responses. MIF acts as a ligand for CD74 receptor and activates the Src-p38 mitogen-activated protein kinase (MAPK) cascade. MIF also elevates the expression of brain-derived neurotrophic factor (BDNF), which contributes to the viability of neurons. Here, we show that MIF is S-nitrosylated by a physiological NO donor. Interestingly, the induction of S-nitrosylation resulted in a loss of MIF activity following stimulation of the Src and p38 MAPK signaling pathways and the induction of BDNF expression. Our results shed light on the pathogenic mechanisms of neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease.

    DOI: 10.1248/bpb.b19-00025

    PubMed

    researchmap

  • Functional analysis of juxta- and intra-membrane domains of murine APP by genome editing in Neuro2a cells. Reviewed International journal

    Nanaka Kaneshiro, Ryosuke Imaoka, Masato Komai, Taku Kashiyama, Takashi Sakurai, Takashi Uehara, Nobumasa Takasugi

    Biochemical and biophysical research communications   501 ( 4 )   1023 - 1028   2018.7

     More details

    Authorship:Last author, Corresponding author   Language:English   Publishing type:Research paper (scientific journal)  

    Amyloid-β precursor protein (APP) correlates with the pathogenesis of certain brain diseases, such as Alzheimer disease (AD). APP is cleaved by several enzymes to produce APP metabolites, including the amyloid beta peptide (Aβ), which accumulates in the brain of AD patients. However, the exact functions of APP metabolites remain elusive. In this study, using genome editing technology, we mutated juxta- and intra-membrane domains of murine APP in the mouse neuroblastoma cell line, Neuro2a. We identified several clones that expressed characteristic patterns of APP metabolites. Mutations in juxta- (deletion 673A), and intra-membrane (deletion 705-6LM) domains of APP, decreased overall levels of APP metabolites or decreased the level of α-secretase-cleaved carboxy-terminal fragment (αCTF), respectively. APP is known to influence neuronal differentiation; therefore, we used theses clones to dissect the function of APP metabolites during neuronal differentiation. One clone (CA), which expressed reduced levels of both FL-APP and αCTF, showed increased expression of the neuronal marker, β3-tubulin, and enhanced retinoic acid (RA)-induced neurite outgrowth. In contrast, a clone that expressed FL-APP, but was devoid of αCTF (CE), showed comparable expression of β3-tubulin and neurite outgrowth compared with normal Neuro2a cells. These data indicate that FL-APP is a suppressor of neurite outgrowth. Our data suggest a novel regulatory function of juxta- and intra-membrane domains on the metabolism and function of APP.

    DOI: 10.1016/j.bbrc.2018.05.102

    PubMed

    researchmap

  • TMEM30A is a candidate interacting partner for the β-carboxyl-terminal fragment of amyloid-β precursor protein in endosomes. Reviewed International journal

    Nobumasa Takasugi, Runa Araya, Yuji Kamikubo, Nanaka Kaneshiro, Ryosuke Imaoka, Hao Jin, Taku Kashiyama, Yoshie Hashimoto, Masaru Kurosawa, Takashi Uehara, Nobuyuki Nukina, Takashi Sakurai

    PloS one   13 ( 8 )   e0200988   2018

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)  

    Although the aggregation of amyloid-β peptide (Aβ) clearly plays a central role in the pathogenesis of Alzheimer's disease (AD), endosomal traffic dysfunction is considered to precede Aβ aggregation and trigger AD pathogenesis. A body of evidence suggests that the β-carboxyl-terminal fragment (βCTF) of amyloid-β precursor protein (APP), which is the direct precursor of Aβ, accumulates in endosomes and causes vesicular traffic impairment. However, the mechanism underlying this impairment remains unclear. Here we identified TMEM30A as a candidate partner for βCTF. TMEM30A is a subcomponent of lipid flippase that translocates phospholipids from the outer to the inner leaflet of the lipid bilayer. TMEM30A physically interacts with βCTF in endosomes and may impair vesicular traffic, leading to abnormally enlarged endosomes. APP traffic is also concomitantly impaired, resulting in the accumulation of APP-CTFs, including βCTF. In addition, we found that expressed BACE1 accumulated in enlarged endosomes and increased Aβ production. Our data suggested that TMEM30A is involved in βCTF-dependent endosome abnormalities that are related to Aβ overproduction.

    DOI: 10.1371/journal.pone.0200988

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0002-6059-2082

  • Consecutive Analysis of BACE1 Function on Developing and Developed Neuronal Cells Reviewed

    Yuji Kamikubo, Nobumasa Takasugi, Kazue Niisato, Yoshie Hashimoto, Takashi Sakurai

    JOURNAL OF ALZHEIMERS DISEASE   56 ( 2 )   641 - 653   2017

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:IOS PRESS  

    The amyloid-beta protein precursor (A beta PP) is cleaved by a transmembrane protease termed beta-site A beta PP cleavage enzyme (BACE1), which is being explored as a target for therapy and prevention of Alzheimer's disease (AD). Although genetic deletion of BACE1 results in abolished amyloid pathology in AD model mice, it also results in neurodevelopmental phenotypes such as hypomyelination and synaptic loss, observed in schizophrenia and autism-like phenotype. These lines of evidence indicate that the inhibition of BACE1 causes adverse side effects during the neurodevelopmental stage. However, the effects of the inhibition of BACE1 activity on already developed neurons remain unclear. Here, we utilized hippocampal slice cultures as an ex vivo model that enabled continuous and long-term analysis for the effect of BACE1 inhibition on neuronal circuits and synapses. Temporal changes in synaptic proteins in hippocampal slices indicated acute synaptic loss, followed by synapse formation and maintenance phases. Long-term BACE1 inhibition in the neurodevelopmental stage caused the loss of synaptic proteins but failed to alter synaptic proteins in the already developed maintenance stage. These data indicate that BACE1 function on synapses is dependent on synaptic developmental stages, and our study provides a useful model to observe the long-term effect of BACE1 activity in the brain, and to evaluate adverse effects of BACE inhibitors.

    DOI: 10.3233/JAD-160806

    Web of Science

    PubMed

    researchmap

  • Modulation of Unfolded Protein Response by Methylmercury. Reviewed

    Hideki Hiraoka, Kengo Nakahara, Yuki Kaneko, Shiori Akiyama, Kosaku Okuda, Takao Iwawaki, Masatake Fujimura, Yoshito Kumagai, Nobumasa Takasugi, Takashi Uehara

    Biological & pharmaceutical bulletin   40 ( 9 )   1595 - 1598   2017

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Methylmercury (MeHg) results in cell death through endoplasmic reticulum (ER) stress. Previously, we reported that MeHg induces S-mercuration at cysteine 383 or 386 in protein disulfide isomerase (PDI), and this modification induces the loss of enzymatic activity. Because PDI is a key enzyme for the maturation of nascent protein harboring a disulfide bond, the disruption in PDI function by MeHg results in ER stress via the accumulation of misfolded proteins. However, the effects of MeHg on unfolded protein response (UPR) sensors and their signaling remain unclear. In the present study, we show that UPR is regulated by MeHg. We found that MeHg specifically attenuated inositol-requiring enzyme 1α (IRE1α)-x-box binding protein 1 (XBP1) branch, but not the protein kinase RNA-like endoplasmic reticulum kinase (PERK) and activating transcriptional factor 6 (ATF6) branches. Treatment with GSK2606414, a specific PERK inhibitor, significantly inhibited MeHg-induced cell death. These findings suggest that MeHg exquisitely regulates UPR signaling involved in cell death.

    DOI: 10.1248/bpb.b17-00359

    PubMed

    researchmap

  • Synthetic ceramide analogues increase amyloid-beta 42 production by modulating gamma-secretase activity Reviewed

    Nobumasa Takasugi, Tomoki Sasaki, Mitsuru Shinohara, Takeshi Iwatsubo, Taisuke Tomita

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   457 ( 2 )   194 - 199   2015.2

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    gamma-Secretase cleaves amyloid beta-precursor protein (APP) to generate amyloid-beta peptide (A beta), which is a causative molecule of Alzheimer disease (AD). The C-terminal length of A beta, which is determined by gamma-secretase activity, determines the aggregation and deposition profiles of A beta, thereby affecting the onset of AD. In this study, we found that the synthetic ceramide analogues DL-threo-1-phenyl-2-decanoylami-no-3-morpholino-1-propanol (PDMP) and (1S,2R-D-erythro-2-N-myristoylamino)-1-phenyl-1-propanol (DMAPP) modulated gamma-secretase-mediated cleavage to increase A beta 42 production. Unexpectedly, PDMP and DMAPP upregulated A beta 42 production independent of alteration of ceramide metabolism. Our results propose that synthetic ceramide analogues function as novel gamma-secretase modulators that increase A beta 42, and this finding might lead to the understanding of the effect of the lipid environment on gamma-secretase activity. (C) 2014 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2014.12.087

    Web of Science

    PubMed

    researchmap

  • FTY720/Fingolimod, a sphingosine analogue, reduces beta-amyloid production in neurons Reviewed

    Nobumasa Takasugi, Tomoki Sasaki, Taisuke Tomita, Takeshi Iwatsubo

    Alzheimer's & Dementia   9 ( 4 )   P184   2013.7

     More details

    Authorship:Lead author   Publisher:Elsevier {BV}  

    DOI: 10.1016/j.jalz.2013.05.311

    researchmap

  • FTY720/Fingolimod, a Sphingosine Analogue, Reduces Amyloid-beta Production in Neurons Reviewed

    Nobumasa Takasugi, Tomoki Sasaki, Ihori Ebinuma, Satoko Osawa, Hayato Isshiki, Koji Takeo, Taisuke Tomita, Takeshi Iwatsubo

    PLOS ONE   8 ( 5 )   2013.5

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:PUBLIC LIBRARY SCIENCE  

    Sphingosine-1-phosphate (S1P) is a pluripotent lipophilic mediator working as a ligand for G-protein coupled S1P receptors (S1PR), which is currently highlighted as a therapeutic target for autoimmune diseases including relapsing forms of multiple sclerosis. Sphingosine related compounds, FTY720 and KRP203 known as S1PR modulators, are phosphorylated by sphingosine kinase 2 (SphK2) to yield the active metabolites FTY720-P and KRP203-P, which work as functional antagonists for S1PRs. Here we report that FTY720 and KRP203 decreased production of Amyloid-beta peptide (A beta), a pathogenic proteins causative for Alzheimer disease (AD), in cultured neuronal cells. Pharmacological analyses suggested that the mechanism of FTY720-mediated A beta decrease in cells was independent of known downstream signaling pathways of S1PRs. Unexpectedly, 6-days treatment of APP transgenic mice with FTY720 resulted in a decrease in A beta 40, but an increase in A beta 42 levels in brains. These results suggest that S1PR modulators are novel type of regulators for A beta metabolisms that are active in vitro and in vivo.

    DOI: 10.1371/journal.pone.0064050

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0002-6059-2082

  • BACE1 Activity Is Modulated by Cell-Associated Sphingosine-1-Phosphate Reviewed

    Nobumasa Takasugi, Tomoki Sasaki, Kunimichi Suzuki, Satoko Osawa, Hayato Isshiki, Yukiko Hori, Naoaki Shimada, Takuya Higo, Satoshi Yokoshima, Tohru Fukuyama, Virginia M. -Y. Lee, John Q. Trojanowski, Taisuke Tomita, Takeshi Iwatsubo

    JOURNAL OF NEUROSCIENCE   31 ( 18 )   6850 - 6857   2011.5

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:SOC NEUROSCIENCE  

    Sphingosine kinase (SphK) 1 and 2 phosphorylate sphingosine to generate sphingosine-1-phosphate (S1P), a pluripotent lipophilic mediator implicated in a variety of cellular events. Here we show that the activity of beta-site APP cleaving enzyme-1 (BACE1), the rate-limiting enzyme for amyloid-beta peptide (A beta) production, is modulated by S1P in mouse neurons. Treatment by SphK inhibitor, RNA interference knockdown of SphK, or overexpression of S1P degrading enzymes decreased BACE1 activity, which reduced A beta production. S1P specifically bound to full-length BACE1 and increased its proteolytic activity, suggesting that cellular S1P directly modulates BACE1 activity. Notably, the relative activity of SphK2 was upregulated in the brains of patients with Alzheimer&apos;s disease. The unique modulatory effect of cellular S1P on BACE1 activity is a novel potential therapeutic target for Alzheimer&apos;s disease.

    DOI: 10.1523/JNEUROSCI.6467-10.2011

    Web of Science

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0002-6059-2082

  • A beta 42 overproduction associated with structural changes in the catalytic pore of gamma-secretase - Common effects of Pen-2 N-terminal elongation and fenofibrate Reviewed

    Noriko Isoo, Chihiro Sato, Hiroyuki Miyashita, Mitsuru Shinohara, Nobumasa Takasugi, Yuichi Morohashi, Shoji Tsuji, Taisuke Tomita, Takeshi Iwatsubo

    JOURNAL OF BIOLOGICAL CHEMISTRY   282 ( 17 )   12388 - 12396   2007.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    gamma-Secretase is an atypical aspartyl protease that cleaves amyloid beta-precursor protein to generate A beta peptides that are causative for Alzheimer disease. gamma-Secretase is a multimeric membrane protein complex composed of presenilin ( PS), nicastrin, Aph-1, and Pen-2. Pen-2 directly binds to transmembrane domain 4 of PS and confers proteolytic activity on gamma-secretase, although the mechanism of activation and its role in catalysis remain unknown. Here we show that an addition of amino acid residues to the N terminus of Pen-2 specifically increases the generation of A beta 42, the longer and more aggregable species of A beta. The effect of the N- terminal elongation of Pen- 2 on A beta 42 generation was independent of the amino acid sequences, the expression system and the presenilin species. In vitro gamma-secretase assay revealed that Pen-2 directly affects the A beta 42- generating activity of gamma-secretase. The elongation of Pen-2N terminus caused a reduction in the water accessibility of the luminal side of the catalytic pore of PS1 in a similar manner to that caused by an A beta 42- raising gamma-secretase modulator, fenofibrate, as determined by substituted cysteine accessibility method. These data suggest a unique mechanism of A beta 42 overproduction associated with structural changes in the catalytic pore of presenilins caused commonly by the N-terminal elongation of Pen-2 and fenofibrate.

    DOI: 10.1074/jbc.M611549200

    Web of Science

    PubMed

    researchmap

  • Aph-1 contributes to the stabilization and trafficking of the gamma-secretase complex through mechanisms involving intermolecular and intramolecular interactions Reviewed

    M Niimura, N Isoo, N Takasugi, M Tsuruoka, K Ui- Tei, K Saigo, Y Morohashi, T Tomita, T Iwatsubo

    JOURNAL OF BIOLOGICAL CHEMISTRY   280 ( 13 )   12967 - 12975   2005.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    gamma-Secretase cleaves type I transmembrane proteins, including beta-amyloid precursor protein and Notch, and requires the formation of a protein complex comprised of presenilin, nicastrin, Aph-1, and Pen-2 for its activity. Aph-1 is implicated in the stabilization of this complex, although its precise mechanistic role remains unknown. Substitution of the first glycine within the transmembrane GXXXG motif of Aph-1 causes a loss-of-function phenotype in Caenorhabditis elegans. Here, using an untranslated region-targeted RNA interference/rescue strategy in Drosophila Schneider 2 cells, we show that Aph-1 contributes to the assembly of the gamma-secretase complex by multiple mechanisms involving intermolecular and intramolecular interactions depending on or independent of the conserved glycines. Aph-1 binds to nicastrin forming an early subcomplex independent of the conserved glycines within the endoplasmic reticulum. Certain mutations in the conserved GXXXG motif affect the interaction of the Aph-1(.)nicastrin subcomplex with presenilin that mediates trafficking of the presenilin(.)Aph-1(.)nicastrin tripartite complex to the Golgi. The same mutations decrease the stability of Aph-1 polypeptides themselves, possibly by affecting intramolecular associations through the transmembrane domains. Our data suggest that the proper assembly of the Aph-1(.)nicastrin subcomplex with presenilin is the prerequisite for the trafficking as well as the enzymatic activity of the gamma-secretase complex and that Aph-1 functions as a stabilizing scaffold in the assembly of this complex.

    DOI: 10.1074/jbc.M409829200

    Web of Science

    PubMed

    researchmap

  • Functional analysis of the presenilin complex and gamma-secretase activity Reviewed

    T Tomita, N Takasugi, M Tsuruoka, M Niimura, Hayashi, I, Y Takahashi, Y Morohashi, N Isoo, S Tanaka, C Sato, T Iwatsubo

    Molecular Neurobiology of Alzheimer Disease and Related Disorders   84 - 93   2004

     More details

    Language:English   Publishing type:Research paper (international conference proceedings)   Publisher:KARGER  

    Web of Science

    researchmap

  • The role of presenilin cofactors in the γ-secratase complex Reviewed

    Nobumasa Takasugi, Taisuke Tomita, Ikuo Hayashi, Makiko Tsuruoka, Manabu Niimura, Yasuko Takahashi, Gopal Thinakaran, Takeshi Iwatsubo

    Nature   422 ( 6930 )   438 - 441   2003.3

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:6930  

    Mutations in presenilin genes account for the majority of the cases of the familial form of Alzheimer's disease (FAD). Presenilin is essential for γ-secretase activity, a proteolytic activity involved in intramembrane cleavage of Notch and β-amyloid precursor protein (βAPP). Cleavage of βAPP by FAD mutant presenilin results in the overproduction of highly amyloidogenic amyloid β42 peptides. γ-Secretase activity requires the formation of a stable, high-molecular-mass protein complex that, in addition to the endoproteolysed fragmented form of presenilin, contains essential cofactors including nicastrin, APH-1 (refs 15-18) and PEN-2 (refs 16, 19). However, the role of each protein in complex formation and the generation of enzymatic activity is unclear. Here we show that Drosophila APH-1 (Aph-1) increases the stability of Drosophila presenilin (Psn) holoprotein in the complex. Depletion of PEN-2 by RNA interference prevents endoproteolysis of presenilin and promotes stabilization of the holoprotein in both Drosophila and mammalian cells, including primary neurons. Co-expression of Drosophila Pen-2 with Aph-1 and nicastrin increases the formation of Psn fragments as well as γ-secretase activity. Thus, APH-1 stabilizes the presenilin holoprotein in the complex, whereas PEN-2 is required for endoproteolytic processing of presenilin and conferring γ-secretase activity to the complex.

    DOI: 10.1038/nature01506

    Scopus

    PubMed

    CiNii Article

    researchmap

    Other Link: http://orcid.org/0000-0002-0075-5943

  • The mechanism of γ-secretase activities through high molecular weight complex formation of presenilins is conserved in Drosophila melanogaster and mammals Reviewed

    Nobumasa Takasugi, Yasuko Takahashi, Yuichi Morohashi, Taisuke Tomita, Takeshi Iwatsubo

    Journal of Biological Chemistry   277 ( 51 )   50198 - 50205   2002.12

     More details

    Authorship:Lead author   Language:English   Publishing type:Research paper (scientific journal)   Publisher:51  

    Mutations in presenilin 1 (PS1) and PS2 genes contribute to the pathogenesis of early onset familial Alzheimer's disease by increasing secretion of the pathologically relevant Aβ42 polypeptides. PS genes are also implicated in Notch signaling through proteolytic processing of the Notch receptor in Caenorhabditis elegans, Drosophila melanogaster, and mammals. Here we show that Drosophila PS (Psn) protein undergoes endoproteolytic cleavage and forms a stable high molecular weight (HMW) complex in Drosophila S2 or mouse neuro2a (N2a) cells in a similar manner to mammalian PS. The loss-of-function recessive point mutations located in the C-terminal region of Psn, that cause an early pupal-lethal phenotype resembling Notch mutant in vivo, disrupted the HMW complex formation, and abolished γ-secretase activities in cultured cells. The overexpression of Psn in mouse embryonic fibroblasts lacking PS1 and PS2 genes rescued the Notch processing. Moreover, disruption of the expression of Psn by double-stranded RNA-mediated interference completely abolished the γ-secretase activity in S2 cells. Surprisingly, γ-secretase activity dependent on wild-type Psn was associated with a drastic overproduction of Aβ1-42 from human βAPP in N2a cells, but not in S2 cells. Our data suggest that the mechanism of γ-secretase activities through formation of HMW PS complex, as well as its abolition by loss-of-function mutations located in the C terminus, are highly conserved features in Drosophila and mammals.

    DOI: 10.1074/jbc.M205352200

    Scopus

    PubMed

    CiNii Article

    researchmap

    Other Link: http://orcid.org/0000-0002-0075-5943

  • Molecular cloning and characterization of CALP/KChIP4, a novel EF-hand protein interacting with presenilin 2 and voltage-gated potassium channel subunit Kv4 Reviewed

    Y Morohashi, N Hatano, S Ohya, R Takikawa, T Watabiki, N Takasugi, Y Imaizumi, T Tomita, T Iwatsubo

    JOURNAL OF BIOLOGICAL CHEMISTRY   277 ( 17 )   14965 - 14975   2002.4

     More details

    Language:English   Publishing type:Research paper (scientific journal)   Publisher:AMER SOC BIOCHEMISTRY MOLECULAR BIOLOGY INC  

    Presenilin (PS) genes linked to early-onset familial Alzheimer's disease encode polytopic membrane proteins that are presumed to constitute the catalytic subunit of gamma-secretase, forming a high molecular weight complex with other proteins. During our attempts to identify binding partners of PS2, we cloned CALP (calsenilin-like protein)/KChIP4, a novel member of calsenilin/ KChIP protein family that interacts with the C-terminal region of PS. Upon co-expression in cultured cells, CALP was directly bound to and co-localized with PS2 in endoplasmic reticulum. Overexpression of CALP did not affect the metabolism or stability of PS complex, and gamma-cleavage of betaAPP or Notch site 3 cleavage was not altered. However, co-expression of CALP and a voltage-gated potassium channel subunit Kv4.2 reconstituted the features of A-type K+ currents and CALP directly bound Kv4.2, indicating that CALP functions as KChIPs that are known as components of native Kv4 channel complex. Taken together, CALP/KChIP4 is a novel EF-hand protein interacting with PS as well as with Kv4 that may modulate functions of a subset of membrane proteins in brain.

    DOI: 10.1074/jbc.M200897200

    Web of Science

    PubMed

    researchmap

  • Presenilin and amyloidogenesis: A structure-function relationship study on presenilin 2 Reviewed

    T Iwatsubo, T Tomita, T Watabiki, R Takikawa, Y Morohashi, N Takasugi

    MAPPING THE PROGRESS OF ALZHEIMER'S AND PARKINSON'S DISEASE   51   65 - 71   2002

     More details

    Language:English   Publishing type:Research paper (international conference proceedings)   Publisher:KLUWER ACADEMIC/PLENUM PUBL  

    Web of Science

    researchmap

  • The First Proline of PALP Motif at the C Terminus of Presenilins is Obligatory for Stabilization, Complex Formation, and γ-Secretase Activities of Presenilins Reviewed

    Taisuke Tomita, Tomonari Watabiki, Rie Takikawa, Yuichi Morohashi, Nobumasa Takasugi, Raphael Kopan, Bart De Strooper, Takeshi Iwatsubo

    Journal of Biological Chemistry   276 ( 35 )   33273 - 33281   2001.8

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Mutations in presenilin (PS) genes cause early-onset familial Alzheimer's disease by increasing production of the amyloidogenic form of amyloid β peptides ending at residue 42 (Aβ42). PS is an evolutionarily conserved multipass transmembrane protein, and all known PS proteins contain a proline-alanine-leucine-proline (PALP) motif starting at proline (P) 414 (amino acid numbering based on human PS2) at the C terminus. Furthermore, missense mutations that replace the first proline of PALP with leucine (P414L) lead to a loss-of-function of PS in Drosophila melanogaster and Caenorhabditis elegans. To elucidate the roles of the PALP motif in PS structure and function, we analyzed neuro2a as well as PS1/2 null fibroblast cell lines transfected with human PS harboring mutations at the PALP motif. P414L mutation in PS2 (and its equivalent in PS1) abrogated stabilization, high molecular weight complex formation, and entry to Golgi/trans-Golgi network of PS proteins, resulting in failure of Aβ42 overproduction on familial Alzheimer's disease mutant basis as well as of site-3 cleavage of Notch. These data suggest that the first proline of the PALP motif plays a crucial role in the stabilization and formation of the high molecular weight complex of PS, the latter being the active form with intramembrane proteolytic activities.

    DOI: 10.1074/jbc.M011152200

    Scopus

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0002-0075-5943

  • C terminus of presenilin is required for overproduction of amyloidogenic Aβ342 through stabilization and endoproteolysis of presenilin Reviewed

    Taisuke Tomita, Rie Takikawa, Akihiko Koyama, Yuichi Morohashi, Nobumasa Takasugi, Takaomi C. Saido, Kei Maruyama, Takeshi Iwatsubo

    Journal of Neuroscience   19 ( 24 )   10627 - 10634   1999.12

     More details

    Language:English   Publishing type:Research paper (scientific journal)  

    Mutations in presenilin (PS) genes cause early onset familial Alzheimer's disease (FAD) by increasing production of the amyloidogenic form of amyloid β peptides ending at residue 42 (Aβ42). To identify a PS subdomain responsible for overproduction of Aβ42, we analyzed neuro2a cell lines expressing modified forms of PS2 that harbor an N1411 FAD mutation. Deletion or addition of amino acids at the C terminus and Ile448 substitution in PS2 with the N1411 FAD mutation abrogated the increase in Aβ42 secretion, and Aβ42 overproduction was de- pendent on the stabilization and endoproteolysis of PS2. The same C-terminal modifications in PS1 produced similar effects. Hence, we suggest that the C terminus of PS plays a crucial role in the overproduction of Aβ42 through stabilization of endoproteolytic PS derivatives and that these derivatives may be the pathologically active species of PS that cause FAD.

    DOI: 10.1523/jneurosci.19-24-10627.1999

    Scopus

    PubMed

    researchmap

    Other Link: http://orcid.org/0000-0002-0075-5943

▼display all

MISC

  • The analysis of endosomal proteins, which specifically interact with APP-beta CTF

    Nobumasa Takasugi, Runa Araya, Yuji Kamikubo, Takashi Sakurai

    JOURNAL OF PHARMACOLOGICAL SCIENCES   124   153P - 153P   2014

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:JAPANESE PHARMACOLOGICAL SOC  

    Web of Science

    researchmap

  • スフィンゴシンキナーゼ/スフィンゴシン-1リン酸シグナリングによるβセクレターゼ活性制御機構の解析

    高杉展正, 富田泰輔, 岩坪威

    日本薬理学会関東部会プログラム・要旨集   126th   2012

  • APP分子内に存在する新規γ-secretase活性制御領域の同定と解析

    佐々木朝輝, 高杉展正, 大沢智子, 富田泰輔, 富田泰輔, 岩坪威, 岩坪威

    Dementia Japan   24 ( 3 )   2010

  • Pen-2のN末端の延長によりAβ42産生上昇を来す機構

    磯尾 紀子, 佐藤 千尋, 宮下 紘幸, 篠原 充, 高杉 展正, 諸橋 雄一, 辻 省次, 富田 泰輔, 岩坪 威

    臨床神経学   47 ( 12 )   1077 - 1077   2007.12

     More details

    Language:Japanese   Publisher:(一社)日本神経学会  

    J-GLOBAL

    researchmap

  • Alzheimer病の分子メカニズム 1) Aβ産生の分子メカニズム

    高杉展正, 富田泰輔, 岩坪威

    医学のあゆみ   206 ( 3 )   2003

  • γセクレターゼ複合体の形成と機能-presenilin cofactorsの役割

    岩坪威, 高杉展正, 富田泰輔

    日本痴呆学会誌   17 ( 1 )   2003

  • Functional analysis of presenilin complex and gamma-secretase activity

    T Tomita, Y Takahashi, Hayashi, I, Y Morohashi, S Tanaka, N Isoo, T Watabiki, M Tsuruoka, N Takasugi, T Iwatsubo

    NEUROBIOLOGY OF AGING   23 ( 1 )   S276 - S277   2002.7

     More details

    Language:English   Publishing type:Research paper, summary (international conference)   Publisher:ELSEVIER SCIENCE INC  

    Web of Science

    researchmap

  • ショウジョウバエプレセニリン(Psn)のγ-セクレターゼ活性及び高分子量複合体形成機構に関する研究

    高杉展正, 鶴岡牧子, 田中さやか, 磯尾紀子, 諸橋雄一, 富田泰輔, 岩坪威

    生化学   74 ( 8 )   2002

  • γセクレターゼによる膜内配列切断におけるプレセニリン複合体の役割

    富田泰輔, 高橋宴子, 林幾雄, 鶴岡牧子, 田中さやか, 磯尾紀子, 諸橋雄一, 高杉展正, 岩坪威

    生化学   74 ( 8 )   2002

  • 非ステロイド系抗炎症剤のγ-secretase阻害効果 in vitro γ-secretase活性測定系を用いた検討

    高橋宴子, 林幾雄, 諸橋雄一, 高杉展正, 富田泰輔, 岩坪威

    生化学   74 ( 8 )   2002

  • 家族性アルツハイマー病原因遺伝子プレセニリンとF-box蛋白

    富田泰輔, 綿引智成, 瀧川里絵, 諸橋雄一, 高杉展正, 岩坪威

    日本薬学会年会要旨集   121st ( 1 )   2001

  • Presenilinとg-secretase

    富田泰輔, 高橋宴子, 綿引智成, 瀧川里絵, 諸橋雄一, 高杉展正, 岩坪威

    日本痴呆学会誌   15 ( 2 )   2001

  • 新規F-box蛋白SEL-10Lのプレセニリン代謝における役割

    綿引智成, 富田泰輔, 高杉展正, 瀧川里絵, 諸橋雄一, 田中啓二, GREENWALD I, 岩坪威

    日本薬学会年会要旨集   121st ( 4 )   2001

  • 新規プレセニリン2C末端結合蛋白CALPのクローニングとその性状解析

    諸橋雄一, 富田泰輔, 瀧川里絵, 高杉展正, 岩坪威

    生化学   72 ( 8 )   2000

▼display all

Industrial property rights

  • アルツハイマー病予防治療薬のスクリーニング法

    高杉 展正, 発明者, 櫻井 隆, 発明者, 清水 瑠奈, 発明者

     More details

    Applicant:学校法人順天堂

    Application no:特願2014-132309(P2014-132309)  Date applied:2014.6.27

    Announcement no:特開特開2016-11849(P2016-11849A)  Date announced:2016.1.21

    researchmap

Awards

  • 生物学研究奨励賞

    2023.10   公益財団法人両備檉園記念財団  

     More details

Research Projects

  • 小胞輸送障害の定量的・網羅的解析法の確立によるアルツハイマー病治療薬の探索

    Grant number:20K07014  2020.04 - 2023.03

    日本学術振興会  科学研究費助成事業 基盤研究(C)  基盤研究(C)

    高杉 展正

      More details

    Grant amount:\4290000 ( Direct expense: \3300000 、 Indirect expense:\990000 )

    アルツハイマー病(AD)は進行性の認知機能の低下を主訴とする神経変性疾患である.近年,患者脳に蓄積するアミロイドβ(Aβ)を標的とした抗体療法が米国で承認されるなど,AD患者脳でAβが蓄積し毒性を持つことがAD発症原因であるとする「アミロイド仮説」が強く支持され創薬研究が進められている.
    一方,Aβ蓄積とは独立した発症機序の存在も示唆されており,病態はより複雑であると考えられ,単一の標的に特化した既知の治療薬の薬効発現は確認しづらく,抗Aβ抗体療法も根本的治療法としてはまだ確立されていないのが現状である.そこでAD病態を正確に把握するため,アミロイド仮説を補完する発症メカニズムの解明と薬物標的の同定が必要とされている.
    これまでにADの最初期の病態である輸送小胞(エンドソーム)の機能異常とその肥大化に注目し,小胞輸送障害がAD発症の端緒であるとする「交通渋滞仮説」が提唱されていたが,そのメカニズムは不明であった.そこで当研究グループはAD関連遺伝子APPの病的代謝物でありAβの前駆体でもあるβCTFに注目した.βCTFは小胞輸送障害を誘導することが知られており,我々はその輸送小胞内での結合パートナーとしてTMEM30Aを同定していた.
    TMEM30Aは脂質二重膜内での脂質輸送に関与し,小胞輸送を制御するリピッドフリッパーゼの構成成分である.本年度は動物・細胞をもちいたAD病態モデルにおいてβCTFの蓄積がリピッドフリッパーゼの形成・活性を低下させることを明らかにした.さらにTMEM30Aに由来するβCTF結合性のペプチドT-RAPを同定し,本ペプチドが小胞輸送障害を改善できることを示し,科学誌に発表した.
    本研究成果からAD発症機構の解明,及び新たな治療標的の同定につながることが期待される.

    researchmap

  • GPCR-Amyloid precursor protein supercomplex regulate neural function

    Grant number:20K07765  2020.04 - 2023.03

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research  Grant-in-Aid for Scientific Research (C)

    上窪 裕二, 高杉 展正

      More details

    Grant amount:\4290000 ( Direct expense: \3300000 、 Indirect expense:\990000 )

    膜タンパク質を標的とした構造と機能解析技術の進歩に伴い、Gタンパク質共役型受容体(GPCR)をはじめとする細胞膜タンパク質間の複合体形成と機能的な相互作用が明らかとなりつつある。膜タンパク質が形成する巨大な複合体は、膜タンパク質の本質であり、その実態を理解することは生命科学の発展に大きく寄与すると考えられる。本研究では多種類の膜タンパク質からなる巨大な複合体(超複合体)の形成とその機能を明らかにするため、アルツハイマー病(AD)の原因の1つであるアミロイドベータ(Aβ)の前駆タンパク質 (Amyloid precursor protein; APP)と神経伝達に関わるGPCR複合体が形成する複合体に注目する。
    先行研究によってAPPの細胞外ドメインの一部が異種GPCR複合体を形成するB型ガンマアミノ酪酸(GABA)受容体(GABAB受容体)と相互作用することが報告された。代表者らはGABAB受容体をはじめとする神経伝達に関わるGPCRを発現する細胞株を作製し、APPおよび部分ペプチドとGPCRの相互作用について検討を行った。検討の結果、GABAB受容体をテトラサイクリン誘導性に発現する細胞株ではAPPとの相互作用は確認できなかった。そこで研究代表者らは、これらの相互作用を安定して評価するためにGABAB受容体を恒常的に発現する細胞株を作製した。さらに、ウイルスベクターによる遺伝子発現方法を改良し、細胞内カルシウムとcAMPをライブセル・イメージングにて評価できる測定系を確立した。申請者らはAPPと他のGPCRの複合体形成についてさらに解析を進め、蛍光ライブセル・イメージングによる評価を行った。

    researchmap

  • APP-BACE1結合仲介分子によるアミロイドβ産生制御機構の解明

    Grant number:19K06929  2019.04 - 2022.03

    日本学術振興会  科学研究費助成事業 基盤研究(C)  基盤研究(C)

    樫山 拓, 高杉 展正

      More details

    Grant amount:\4290000 ( Direct expense: \3300000 、 Indirect expense:\990000 )

    アルツハイマー病の病理に中心的役割を果たすアミロイドβ(Aβ)は前駆体タンパク質(APP)がBACE1・γセクレターゼによる2段階切断を受けて産生される。Aβ凝集体からなる老人斑の近傍では、神経突起における小胞輸送障害により変性神経突起が形成される。輸送障害部位ではAPP・BACE1の蓄積が起こり、悪循環的にAβ産生亢進、凝集体の形成促進、変性神経突起形成・神経機能障害が進行すると考えられている。 我々がAPP結合タンパク質として見出したタンパク質Xが老人班周囲に集積することを報告しているが、さらにタンパク質XがBACE1とも結合することを発見した。以上のことから、このタンパク質XがAPPとBACE1の集積を促し、Aβ産生を亢進しているというのではないかという仮説を立てた。タンパク質Xの機能を修飾することでAβ産生を抑制できれば新たなアルツハイマー病治療薬の標的となることが期待される。 アルツハイマー病モデルマウス脳スライス固定標本の免疫染色によりアミロイドプラーク周辺におけるBACE1およびタンパク質Xの集積を確認した。BACE1とタンパク質Xの同時染色に必要な抗体が無いため、抗BACE1-VHH抗体を利用し共局在を検討したが、感度が不十分なため改善が必要である。
    タンパク質XとBACE1の結合部位を明らかにするため、それぞれについて部分欠損変異体を作製し、株化細胞を用いて免疫沈降、および蛍光顕微鏡による共局在を指標に結合を評価した。少なくとも互いの膜貫通ドメイン同士の結合が確認され、加えて細胞外ドメインの複数の結合部位が存在することが示唆された。タンパク質XはBACE1以外に他のタンパク質と複合体を形成することが示されていることから、3者が存在するときの結合様式や振る舞いをさらに検討する必要がある。

    researchmap

  • Lipid flippase is the novel therapeutic target for the Alzheimer disease, which correlate with endocytic traffic impairment.

    Grant number:17K08272  2017.04 - 2020.03

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research  Grant-in-Aid for Scientific Research (C)

    TAKASUGI NOBUMASA

      More details

    Grant amount:\4810000 ( Direct expense: \3700000 、 Indirect expense:\1110000 )

    Although Amyloid beta (Aβ) peptide, which accumulates in Alzheimer disease (AD) brain, is focused attention as a therapeutic target, most clinical trials are failed. Interestingly, endocytic dysfunction is the early pathogenic event by the accumulation of Aβ precursor protein (APP) metabolites, β-carboxyl-terminal fragment (βCTF). We identified TMEM30A, a subcomponent of lipid flippase, as a candidate partner for βCTF. Moreover, stably overexpression of β-site APP cleaving enzyme 1 (BACE1) accumulated βCTF to increase the complex with TMEM30A. Intriguingly, upregulated BACE1 activity reduced both the lipid flippase formation and activity, which was accompanied by the abnormally enlarged endosomes. We confirmed the complex formation of TMEM30A and βCTF, and lipid flippase dysfunction in AD model mice before Aβ deposition. Our results shed light on the unidentified correlation between βCTF and lipid flippase activity and suggest a novel therapeutic strategy for AD.

    researchmap

  • Simultaneous control of the production and degradation of Amyloid be-ta by Sphingosine kinase/sphingosine-1-phosphate signaling

    Grant number:26430059  2014.04 - 2017.03

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (C)  Grant-in-Aid for Scientific Research (C)

    TAKASUGI NOBUMASA, TOMITA Taisuke

      More details

    Grant amount:\5200000 ( Direct expense: \4000000 、 Indirect expense:\1200000 )

    Alzheimer disease (AD)is a progressive neurodegenerative disease. The over-production and aggregation of Amyloid beta (Aβ) peptides in brains are considered to be the major pathogenic event for AD. We identified Sphingosine kinase 2 (SphK2) activity is upregulated in AD, and caused to increase the level of Aβ. These results strongly suggest SphK2 and its product sphingosine-1-phosphate are the therapeutic target for AD.

    researchmap

  • Microdomain-dependent acceleration mechanisms for amyloidogenic processing of amyloid precursor protein induced by endocytic membrane traffic impairment

    Grant number:23300128  2011.04 - 2015.03

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Scientific Research (B)  Grant-in-Aid for Scientific Research (B)

    SAKURAI Takashi, NUKINA Nobuyuki, MURAYAMA Takashi, KASHIYAMA Taku, ARAYA Runa, KAMIKUBO Yuji, TAKASUGI Nobumasa

      More details

    Grant amount:\19760000 ( Direct expense: \15200000 、 Indirect expense:\4560000 )

    Aggregation and deposition of amyloid β peptide (Aβ) in the brain is considered central to the pathogenesis of Alzheimer’s disease (AD). Aβ is produced through a sequential cleavage of the amyloid precursor protein (APP) by β- and γ-secretases. Increasing evidence indicates that membrane microdomains play an important role as a platform for the vesicular transport of APP in the secretory and endocytic pathways and in the amyloidogenic processing of APP. Recently, accumulation of β-secretase-cleaved C-terminal fragment of APP (βCTF), which is the direct precursor of Aβ, was found to cause endosome dysfunction during the early phase of AD that could drive Aβ overproduction. To gain insight into the underlying molecular mechanisms, we examined the functions of endosomal proteins in the APP-containing microdomains. We identified a candidate protein that causes impairment of endocytic membrane traffic through its interactions with βCTF.

    researchmap

  • SPHK/S1Pシグナリングによるβセクレターゼ制御機構の解析

    Grant number:21790061  2009 - 2010

    日本学術振興会  科学研究費助成事業 若手研究(B)  若手研究(B)

    高杉 展正

      More details

    Grant amount:\4290000 ( Direct expense: \3300000 、 Indirect expense:\990000 )

    高齢化社会を迎え、アルツハイマー病(AD)は大きな社会問題となっている。その発症機序には、β及びγセクレターゼによる2段階の切断によりAmyloid precursor protein (APP)から生じるAβ分子の患者脳内での蓄積が深く関与している。申請者らは脂質セカンドメッセンジャーとして機能することが知られるSphingosine-1-phoaphate (S1P)の産生酵素であるSphingosine kinase (SPHK)がβセクレターゼを介してAβ産生機構影響を与えることを見出していた。本年度は細胞内S1Pの量が実際にβセクレターゼ活性に影響して、AD発症に寄与する可能性があるか検討することを主眼として以下の研究を進めた。
    1.S1P代謝酵素によるβセクレターゼ活性制御機構に対する影響の解析
    2.SPHKの過剰発現がAβ産生機構に与える影響の解析
    結果として分子機構の異なるS1P代謝酵素であるSGPL1、SGPP1の発現によりヒト由来のAβおよびβセクレターゼ産物であるβCTFの産生が有意に抑制されることを見出した。またSPHKの一つであるSPHK2の発現は酵素活性依存的にAβ、βCTF産生を増加させることが明らかになった
    これらの結果から、これまでに明らかでなかったSPHK/S1Pシグナル、特に細胞内S1Pの量が神経細胞におけるβセクレターゼの活性を制御していることが示唆された。S1P量は免疫応答、神経細胞へのストレスなどにより影響をうける可能性があることから、AD発症機構に関与していることも考えられる。
    本研究成果から、今後SPHKおよびS1P代謝酵素をターゲットとしたAD根本治療薬の創薬につながることが期待される。

    researchmap

  • Substrate specific modulation of the ....secretase activity by vesicular trafficking

    Grant number:19790049  2007 - 2008

    Japan Society for the Promotion of Science  Grants-in-Aid for Scientific Research Grant-in-Aid for Young Scientists (B)  Grant-in-Aid for Young Scientists (B)

    TAKASUGI Nobumasa

      More details

    Grant amount:\3690000 ( Direct expense: \3300000 、 Indirect expense:\390000 )

    アルツハイマー病(AD)患者脳に蓄積するアミロイドβ(Aβ)蛋白質を産生する責任酵素であるγセクレターゼは、AD の根本的治療法のターゲットとして注目されている。しかしその単純な阻害はNotch などのシグナルの阻害による副作用を誘発する。我々はショウジョウバエ細胞を用い、Notch、Aβ産生を特異的に制御する遺伝子をRNAi 法によるスクリーニングをおこない、候補遺伝子について個々に解析を進めた。本研究成果は副作用のないAD 治療薬の開発・および生体内シグナルの重要な因子であるNotch の制御機構の解析に道筋をつける研究であると考えている。

    researchmap

  • Drosophilaを用いたパーキンソン病、アルツハイマー病の病態に関する研究

    Grant number:02J61410  2002 - 2003

    日本学術振興会  科学研究費助成事業 特別研究員奨励費  特別研究員奨励費

    高杉 展正

      More details

    Grant amount:\1000000 ( Direct expense: \1000000 )

    家族性アルツハイマー病(FAD)病因遺伝子presenilin(PS)の変異によりADが発症する機序として、アミロイドとして蓄積性の高いAβ42ペプチドの産生亢進が報告され、ADの根本的治療法の創薬ターゲットとして注目されている。PSの正常機能については不明な点が多いが、PSはβアミロイド前駆体蛋白(βAPP)や細胞分化に重要な役割を果たすNotch受容体の膜内配列切断を行う新規アスパルチルプロテアーゼγ-secretaseの活性サブユニットである可能性が示唆されている。
    これまでに我々は、断片化したPSは安定化され、高分子量複合体を形成すること、この複合体が活性型γ-secretaseの本態であることを明らかにしてきた。私はγ-secretaseの分子的実態を明らかにすることを目的として、分子遺伝学的解析法の確立されているショウジョウバエを実験系として用い、ショウジョウバエプレセニリン(Psn)の解析を行った。ショウジョウバエ由来シュナイダー(S2)細胞において内因性Psnは断片化、安定化を受け高分子量複合体を形成しており、S2細胞にβAPPのC末端断片(C100)を発現させるとAβが産生され、Psnがγ-secretaseとしての活性を持つことを明らかにした。一方マウス由来N2a細胞にPsnを発現させた場合にも、哺乳類PSと同様に安定化、高分子量複合体を形成し、γ-secretase活性を示した。これらの結果はPSの安定化、高分子量複合体形成機構が遺伝的に保存されており、S2細胞及びショウジョウバエPsnを用いた系がγ-secretase活性を評価するモデルとして有用であることを示している。現在このS2細胞を利用した実験系により、γ-セクレターゼの新たな構成因子候補として同定されたNicastrin、Aph-1、Pen-2について解析している。

    researchmap

▼display all

 

Class subject in charge

  • How Do Medicines Work ? (2023academic year) Fourth semester  - 木1~2

  • Practice in Clinical and Biopharmaceutical Sciences (2023academic year) Third semester  - その他5~9

  • Practice in Clinical and Biopharmaceutical Sciences (2023academic year) Third semester  - その他5~9

  • Practice in Clinical and Biopharmaceutical Sciences (2023academic year) Third semester  - その他5~9

  • Practice in Clinical and Biopharmaceutical Sciences (2023academic year) Third semester  - その他5~9

  • Life Science and Molecular Cell Biology (2023academic year) special  - その他

  • Life Science 1 (2023academic year) special  - その他

  • Pharmacology (2023academic year) special  - その他

  • Pharmacology (2023academic year) special  - その他

  • Practicals: Medicinal Pharmacology (2023academic year) special  - その他

  • Research Projects: Medicinal Pharmacology (2023academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology A I (2023academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology A I (2023academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology A II (2023academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology A II (2023academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology B I (2023academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology B I (2023academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology B II (2023academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology B II (2023academic year) special  - その他

  • Pharmacology 3 (2023academic year) 1st semester  - 金3~4

  • Pharmacology 3 (2023academic year) 1st semester  - 金3~4

  • Pharmacology 4 (2023academic year) Second semester  - 金3~4

  • Pharmacology 4 (2023academic year) Second semester  - 金3~4

  • Pharmacology 5 (2023academic year) Third semester  - 月1~2

  • Pharmacology 5 (2023academic year) Third semester  - 月1~2

  • Pharmacology 6 (2023academic year) Fourth semester  - 月1~2

  • Pharmacology 6 (2023academic year) Fourth semester  - 月1~2

  • Pharmacology B (2023academic year) 1st and 2nd semester  - 金3~4

  • Pharmacology B (2023academic year) 1st and 2nd semester  - 金3~4

  • Pharmacology C (2023academic year) 3rd and 4th semester  - 月1~2

  • Pharmacology C (2023academic year) 3rd and 4th semester  - 月1~2

  • How Do Medicines Work ? (2022academic year) Fourth semester  - 木1~2

  • Practice in Clinical and Biopharmaceutical Sciences (2022academic year) Third semester  - その他5~9

  • Practice in Clinical and Biopharmaceutical Sciences (2022academic year) Third semester  - その他5~9

  • Life Science 1 (2022academic year) special  - その他

  • Pharmacology (2022academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology A I (2022academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology A I (2022academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology A II (2022academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology A II (2022academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology B I (2022academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology B I (2022academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology B II (2022academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology B II (2022academic year) special  - その他

  • Pharmacology 3 (2022academic year) 1st semester  - 金3~4

  • Pharmacology 3 (2022academic year) 1st semester  - 金3~4

  • Pharmacology 4 (2022academic year) Second semester  - 金3~4

  • Pharmacology 4 (2022academic year) Second semester  - 金3~4

  • Pharmacology 5 (2022academic year) Third semester  - 月1~2

  • Pharmacology 5 (2022academic year) Third semester  - 月1~2

  • Pharmacology 6 (2022academic year) Fourth semester  - 月1~2

  • Pharmacology 6 (2022academic year) Fourth semester  - 月1~2

  • How Do Medicines Work ? (2021academic year) Fourth semester  - 木1~2

  • Practice in Clinical and Biopharmaceutical Sciences (2021academic year) Third semester  - その他6~9

  • Practice in Clinical and Biopharmaceutical Sciences (2021academic year) Third semester  - その他6~9

  • Life Science 1 (2021academic year) Prophase  - その他

  • Pharmacology (2021academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology A I (2021academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology A I (2021academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology A II (2021academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology A II (2021academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology B I (2021academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology B I (2021academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology B II (2021academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology B II (2021academic year) special  - その他

  • Basic Practice in Pharmaceutical Sciences (2021academic year) 1-3 semesters  - その他6~9

  • Basic Practice in Pharmaceutical Sciences (2021academic year) 1-3 semesters  - その他6~9

  • Pharmacology 3 (2021academic year) 1st semester  - 金3,金4

  • Pharmacology 3 (2021academic year) 1st semester  - 金3,金4

  • Pharmacology 4 (2021academic year) Second semester  - 金3,金4

  • Pharmacology 4 (2021academic year) Second semester  - 金3,金4

  • Pharmacology 5 (2021academic year) Third semester  - 月1,月2

  • Pharmacology 5 (2021academic year) Third semester  - 月1,月2

  • Pharmacology 6 (2021academic year) Fourth semester  - 月1,月2

  • Pharmacology 6 (2021academic year) Fourth semester  - 月1,月2

  • How Do Medicines Work ? (2020academic year) Fourth semester  - 木1,木2

  • Practice in Clinical and Biopharmaceutical Sciences (2020academic year) Third semester  - その他

  • Practice in Clinical and Biopharmaceutical Sciences (2020academic year) Third semester  - その他

  • Life Science 1 (2020academic year) special  - その他

  • Pharmacology (2020academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology A I (2020academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology A I (2020academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology A II (2020academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology A II (2020academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology B I (2020academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology B I (2020academic year) special  - その他

  • Research Projects and Practicals: Medicinal Pharmacology B II (2020academic year) special  - その他

  • Lecture and Research Projects: Medicinal Pharmacology B II (2020academic year) special  - その他

  • Basic Practice in Pharmaceutical Sciences (2020academic year) special  - その他

  • Basic Practice in Pharmaceutical Sciences (2020academic year) special  - その他

  • Pharmacology 1 (2020academic year) Third semester  - 木5,木6

  • Pharmacology 1 (2020academic year) Third semester  - 木5,木6

  • Pharmacology 2 (2020academic year) Fourth semester  - 木3,木4

  • Pharmacology 2 (2020academic year) Fourth semester  - 木3,木4

  • Pharmacology 3 (2020academic year) 1st semester  - 金3,金4

  • Pharmacology 3 (2020academic year) 1st semester  - 金3,金4

  • Pharmacology 4 (2020academic year) Second semester  - 金3,金4

  • Pharmacology 4 (2020academic year) Second semester  - 金3,金4

  • Pharmacology 7 (2020academic year) Third semester  - 金3,金4

  • Pharmacology 7 (2020academic year) Third semester  - 金3,金4

  • Pharmacology 8 (2020academic year) Fourth semester  - 金3,金4

  • Pharmacology 8 (2020academic year) Fourth semester  - 金3,金4

  • Pharmacology I (2020academic year) 3rd and 4th semester  - 木5,木6

  • Pharmacology II (2020academic year) 1st and 2nd semester  - 金3,金4

  • Pharmacology II (2020academic year) 1st and 2nd semester  - 金3,金4

  • Pharmacology IV (2020academic year) 3rd and 4th semester  - 金3,金4

▼display all