研究者業績

新倉 貴子

ニイクラ タカコ  (Niikura Takako)

基本情報

所属
上智大学 理工学部情報理工学科 教授
学位
博士(薬学)(東京大学)

J-GLOBAL ID
201301051278280653
researchmap会員ID
7000004363

1989年ー1991年 化合物安全性研究所
1991年ー1992年 東燃株式会社
1992年ー1993年 NERC Institute of Virology and Environmental Microbiology (Oxford, UK)
1994年ー1995年 Thomas Jefferson University, School of Medicine (Philadelphia, USA)
1995年ー1997年 Michigan State University, Department of Biochemistry (East Lansing, USA)
1997年ー2007年 慶應義塾大学医学部
2007年ー2009年 Georgetown University, Department of Neurology (Washington DC, USA)
2009年ー2012年 Simon Fraser University, Faculty of Health Sciences (Burnaby, Canada)
2012年ー現在   上智大学理工学部情報理工学科


研究キーワード

 3

論文

 69
  • Y Yamagishi, Y Hashimoto, T Niikura, Nishimoto, I
    PEPTIDES 24(4) 585-595 2003年4月  査読有り
    Humanin (HN) is a secretory peptide that inhibits neurotoxicity by various Alzheimer's disease-relevant insults. We have so far identified that the substitution of Leu9 for Arg nullifies the extracellular secretion of HN. Here we comprehensively investigate the amino acid requirement of HN essential for its secretion and for its neuroprotective function. Intracellulary expressed HN-EGFP (EGFP N-terminally fused with HN) was extracellularly secreted, whereas neither EGFP nor (L9R)HN-EGFP was secreted at all. While Ala substitution of neither residue affected HN secretion, Arg substitution revealed that the two structures-Leu9-Leu11 and Pro 19-Val 20-were essential for the secretion of full-length HN. In the Leu9-Leu11 domain, the Leu10 residue turned out to play a central role in this function, because the Asp substitution of Leu10, but not Leu9 or Leu11, nullified the secretion of HN. Utilizing Ala-scanned HN constructs, we also investigated a comprehensive structure-function relationship for the neuroprotective function of full-length HN, which revealed (i) that Pro3, Ser7, Cys8, Leu9, Leu12, Thr13, Ser14, and Pro19 were essential for this function and (ii) that Ser7 and Leu9 were essential for self-dimerization of HN. These findings indicate that HN has activity similar to a signal peptide, for which the Leu9-Leu11 region, particularly Leu10, functions as a core domain, and suggest that self-dimerization of HN is a process essential for its neuroprotective function. (C) 2003 Elsevier Science Inc. All rights reserved.
  • Takako Niikura, Yuichi Hashimoto, Hirohisa Tajima, Miho Ishizaka, Yohichi Yamagishi, Masaoki Kawasumi, Mikiro Nawa, Kenzo Terashita, Sadakazu Aiso, Ikuo Nishimoto
    European Journal of Neuroscience 17(6) 1150-1158 2003年3月  査読有り
  • Y Hashimoto, O Tsuji, T Niikura, Y Yamagishi, M Ishizaka, M Kawasumi, T Chiba, K Kanekura, M Yamada, E Tsukamoto, K Kouyama, K Terashita, S Aiso, A Lin, Nishimoto, I
    JOURNAL OF NEUROCHEMISTRY 84(4) 864-877 2003年2月  査読有り
    Amyloid precursor protein (APP), the precursor of Abeta, has been shown to function as a cell surface receptor that mediates neuronal cell death by anti-APP antibody. The c-Jun N-terminal kinase (JNK) can mediate various neurotoxic signals, including Abeta neurotoxicity. However, the relationship of APP-mediated neurotoxicity to JNK is not clear, partly because APP cytotoxicity is Abeta independent. Here we examined whether JNK is involved in APP-mediated neuronal cell death and found that: (i) neuronal cell death by antibody-bound APP was inhibited by dominant-negative JNK, JIP-1b and SP600125, the specific inhibitor of JNK, but not by SB203580 or PD98059; (ii) constitutively active (ca) JNK caused neuronal cell death and (iii) the pharmacological profile of caJNK-mediated cell death closely coincided with that of APP-mediated cell death. Pertussis toxin (PTX) suppressed APP-mediated cell death but not caJNK-induced cell death, which was suppressed by Humanin, a newly identified neuroprotective factor which inhibits APP-mediated cytotoxicity. In the presence of PTX, the PTX-resistant mutant of Galpha(o) , but not that of Galpha(i) , recovered the cytotoxic action of APP. These findings demonstrate that JNK is involved in APP-mediated neuronal cell death as a downstream signal transducer of G(o) .
  • H Tajima, T Niikura, Y Hashimoto, Y Ito, Y Kita, K Terashita, K Yamazaki, A Koto, S Aiso, Nishimoto, I
    NEUROSCIENCE LETTERS 324(3) 227-231 2002年5月  査読有り
    An unbiased functional screening with brain cDNA library from an Alzheimer's disease (AD) brain identified a novel 24-residue peptide Humanin (HN), which suppresses AD-related neurotoxicity. As the 1567-base cDNA containing the open reading frame (ORF) of HN is 99% identical to mitochondrial 16S ribosomal RNA as well as registered human mRNA, it was elusive whether HN is produced in vivo. Here, we raised anti-HN antibody and found that long cDNAs containing the ORF of HN (HN-ORF) produced the HN peptide in mammalian cells, dependent on the presence of full-length HN-ORF. Immunoblot analysis detected a 3-kDa protein with HN immunoreactivity in the testis and the colon in 3-week-old mice and in the testis in 12-week-old mice. HN immunoreactivity was also detected in an AD brain, but little in normal brains. This study suggests that HN peptide could be produced in vivo, and would provide a novel insight into the pathophysiology of AD. (C) 2002 Elsevier Science Ireland Ltd. All rights reserved.
  • Y Hashimoto, T Niikura, Y Ito, Y Kita, K Terashita, Nishimoto, I
    JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS 300(3) 736-745 2002年3月  査読有り
    Although it has been established that oxidative stress mediates cytotoxicity by familial Alzheimer's disease (FAD)-linked mutants of presenilin (PS)1 and that pertussis toxin inhibits cytotoxicity by FAD-linked N141I-PS2, it has not been determined whether oxidative stress is involved in cytotoxicity by N141I-PS2 or which pertussis toxin-sensitive proteins mediate the cytotoxicity. Here we report that low expression of N141I-PS2 caused neuronal cell death, whereas low expression of wildtype PS2 did not. Cytotoxicities by low and high expression of N141I-PS2 occurred through dissimilar mechanisms: the former cytotoxicity was blocked by a cell-permeable caspase inhibitor, and the latter was not. Since both mechanisms were sensitive to a cell-permeable antioxidant, we examined potential sources of reactive oxygen species in each mechanism, and found that the caspase inhibitor-sensitive neurotoxicity by N141I-PS2 was likely through NADPH oxidase and the caspase inhibitor-resistant neurotoxicity by N141I-PS2 through xanthine oxidase. Pertussis toxin greatly suppressed both toxic mechanisms by N141I-PS2, and only Galpha(o), a neuron-enriched pertussis toxin-sensitive G protein, was involved in both mechanisms. We therefore conclude that N141I-PS2 is capable of triggering multiple neurotoxic mechanisms, which can be inhibited by the combination of clinically usable inhibitors of NADPH oxidase and xanthine oxidase. This study thus provides a novel insight into the therapeutic intervention of PS2 mutant-associated FAD.
  • Yuichi Hashimoto, Yuko Ito, Erika Arakawa, Yoshiko Kita, Kenzo Terashita, Takako Niikura, Ikuo Nishimoto
    Journal of Neurochemistry 80(3) 426-437 2002年2月  査読有り
  • Yuichi Hashimoto, Takako Niikura, Yuko Ito, Haruka Sudo, Michihiro Hata, Erika Arakawa, Yoichiro Abe, Yoshiko Kita, Ikuo Nishimoto
    The Journal of neuroscience : the official journal of the Society for Neuroscience 21(23) 9235-9245 2001年12月  査読有り
  • S Matsuda, T Yasukawa, Y Homma, Y Ito, T Niikura, T Hiraki, S Hirai, S Ohno, Y Kita, M Kawasumi, K Kouyama, T Yamamoto, JM Kyriakis, Nishimoto, I
    JOURNAL OF NEUROSCIENCE 21(17) 6597-6607 2001年9月  査読有り
    Using a yeast two-hybrid method, we searched for amyloid precursor protein (APP)-interacting molecules by screening mouse and human brain libraries. In addition to known interacting proteins containing a phosphotyrosine-interaction-domain (PID)-Fe65, Fe65L, Fe65L2, X11, and mDab1, we identified, as a novel APP-interacting molecule, a PID-containing isoform of mouse JNK-interacting protein-1 (JIP-1b) and its human homolog IB1, the established scaffold proteins for JNK. The APP amino acids Tyr(682), Asn(684), and Tyr(687) in the G(681)YENPTY(687) region were all essential for APP/JIP-1b interaction, but neither Tyr(653) nor Thr(668) was necessary. APP-interacting ability was specific for this additional isoform containing PID and was shared by both human and mouse homologs. JIP-1b expressed by mammalian cells was efficiently precipitated by the cytoplasmic domain of APP in the extreme Gly(681)-Asn(695) domain-dependent manner. Reciprocally, both full-length wild-type and familial Alzheimer's disease mutant APPs were precipitated by PID-containing JIP constructs. Antibodies raised against the N and C termini of JIP-1b coprecipitated JIP-1b and wild-type or mutant APP in nonneuronal and neuronal cells. Moreover, human JNK1 beta1 formed a complex with APP in a JIP-1b-dependent manner. Confocal microscopic examination demonstrated that APP and JIP-1b share similar subcellular localization in transfected cells. These data indicate that JIP-1b/IB1 scaffolds APP with JNK, providing a novel insight into the role of the JNK scaffold protein as an interface of APP with intracellular functional molecules.
  • Yuichi Hashimoto, Takako Niikura, Hirohisa Tajima, Takashi Yasukawa, Haruka Sudo, Yuko Ito, Yoshiko Kita, Masaoki Kawasumi, Keisuke Kouyama, Manabu Doyu, Gen Sobue, Takashi Koide, Shoji Tsuji, Jochen Lang, Kiyoshi Kurokawa, Ikuo Nishimoto
    Proceedings of the National Academy of Sciences of the United States of America 98(11) 6336-6341 2001年5月  査読有り
  • Y Hashimoto, Y Ito, T Niikura, Z Shao, M Hata, F Oyama, Nishimoto, I
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 283(2) 460-468 2001年5月  査読有り
    We report a novel gene, designated Humanin (HN) cDNA, that suppresses neuronal cell death by K595N/M596L-APP (NL-APP), a mutant causing familial Alzheimer's disease (FAD), termed Swedish mutant, Transfection of neuronal cells with HN cDNA or treatment with the coding HN polypeptide abrogated cytotoxicity by NL-APP, HN suppressed neurotoxicity by A beta1-43 in the absence of N2 supplement, but could not inhibit AP secretion from NL-APP, HN could also protect neuronal cells from death by NL-APP lacking the 41st and 42nd residues of the A beta region, Therefore, HN suppressed neuronal cell death by NL-APP not through inhibition of A beta 42 secretion, but with two targets for its inhibitory action: (i) the intracellular toxic mechanism directly triggered by NL-APP and (ii) neurotoxicity by A beta. HN will contribute to the development of curative therapy of AD, especially as a novel reagent that could mechanistically supplement A beta -production inhibitors. (C) 2001 Academic Press.
  • T Niikura, Y Hashimoto, T Okamoto, Y Abe, T Yasukawa, M Kawasumi, T Hiraki, Y Kita, K Terashita, K Kouyama, Nishimoto, I
    JOURNAL OF NEUROSCIENCE 21(6) 1902-1910 2001年3月  査読有り
    It has been found that insulin-like growth factor I (IGF-I) exerts cytoprotection against A beta amyloid-induced neuronal cell death. Deposits of A beta amyloid are one of the pathological hallmarks of Alzheimer's disease (AD). Here, we examined whether IGF-I exerts protective activity against cell death induced by a familial AD (FAD)-linked mutant of amyloid precursor protein (APP), and we found that IGF-I protected cells from toxicity of FAD-associated V642I mutant of APP in multiple cell systems. IGFBP-3 blocked this action of IGF-I, but not of des( 1-3) IGF-I, which was as active as IGF-I in the presence of IGFBP-3. The data also demonstrated that the IGF-I receptor (IGF-IR) mediates the protective activity of IGF-I. The antagonizing function of the IGF-I/IGF-IR system against V642I-APP, which is further antagonized by IGFBP-3, provides a molecular clue to the understanding of AD pathophysiology and to the establishment of potential therapy for AD.
  • Haruka Sudo, Yuichi Hashimoto, Takako Niikura, Zongjun Shao, Takashi Yasukawa, Yuko Ito, Marina Yamada, Michihiro Hata, Takako Hiraki, Masaoki Kawasumi, Keisuke Kouyama, Ikuo Nishimoto
    Biochemical and Biophysical Research Communications 282(2) 548-556 2001年3月  査読有り
  • Haruka Sudo, Hong Jiang, Takashi Yasukawa, Yuichi Hashimoto, Takako Niikura, Masaoki Kawasumi, Shuji Matsuda, Yuji Takeuchi, Sadakazu Aiso, Masaaki Matsuoka, Yoshitake Murayama, Ikuo Nishimoto
    Molecular and Cellular Neuroscience 16(6) 708-723 2000年12月  査読有り
  • A Hagiwara, Y Hashimoto, T Niikura, Y Ito, K Terashita, Y Kita, Nishimoto, I, K Umezawa
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 278(3) 633-639 2000年11月  査読有り
    Since an apolipoprotein E4 (ApoE4) peptide composed of the low-density lipoprotein (LDL) receptor-related protein (LRP)-binding domain [ApoE4(141-149)(2) or ApoE(141-155)(2)] exerts neurotoxicity in primary newtons and neuronal cell lines, it has been controversial whether these effects are mediated by LRP. Here, we examined whether ApoE4(141-149)(2)-induced toxicity is mediated by LRP in a neuronal cell system where ApoE4 toxicity is mediated by LRP: serum-deprived F11 neuronal cells. In these cells, where ApoE4 exerted toxicity by apoptosis in a manner sensitive to both caspase inhibitors and pertussis toxin (PTX), ApoE4(141-149)(2) also caused cell death by apoptosis but in a caspase-inhibitor-resistant, PTX-resistant manner. ApoE4(141-149)(2)-induced death was not inhibited by antisense oligonucleotides to LRP. Therefore, we conclude that ApoE4(141-149)(2) is able to exert neurotoxicity without involving LRP. (C) 2000 Academic Press.
  • Y Hashimoto, H Jiang, T Niikura, Y Ito, A Hagiwara, K Umezawa, Y Abe, Y Murayama, Nishimoto, I
    JOURNAL OF NEUROSCIENCE 20(22) 8401-8409 2000年11月  査読有り
    The epsilon4 genotype of apolipoprotein E (apoE4) is the most established predisposing factor in Alzheimer's disease (AD); however, it remains unclear how apoE4 contributes to the pathophysiology. Here, we report that the apoE4 protein (ApoE4) evokes apoptosis in neuronal cells through the low-density lipoprotein receptor-related protein (LRP) and heterotrimeric GTPases. We examined neuron/neuroblastoma hybrid F11 cells and found that these cells were killed by 30 mug/ml ApoE4, but not by 30 mg/ml ApoE3. ApoE4-induced death occurred with typical features for apoptosis in time- and dose-dependent manners, and was observed in SH-SY5Y neuroblastomas, but not in glioblastomas or non-neuronal Chinese hamster ovary cells. Activated, but not native, alpha2-macroglobulin suppressed this ApoE4 toxicity. Suppression by the antisense oligonucleotide to LRP and inhibition by low nanomolar concentrations of LRP-associated protein RAP provided evidence for the involvement of LRP. The involvement of heterotrimeric GTPases was demonstrated by the findings that (1) ApoE4-induced death was suppressed by pertussis toxin (PTX), but not by heat-inactivated PTX; and (2) transfection with PTX-resistant mutant cDNAs of G alpha (i) restored the toxicity of ApoE4 restricted by PTX. We thus conclude that one of the neurotoxic mechanisms triggered by ApoE4 is to activate a cell type-specific apoptogenic program involving LRP and the Gi class of GTPases and that the apoE4 gene may play a direct role in the pathogenesis of AD and other forms of dementia.
  • Y Hashimoto, T Niikura, Y Ito, Nishimoto, I
    JOURNAL OF BIOLOGICAL CHEMISTRY 275(44) 34541-34551 2000年11月  査読有り
    We examined a neuronal cell system in which single-cell expression of either familial Alzheimer's disease (FAD) gene V642I-APP or K595N/M596L-APP (NL-APP) in an inducible plasmid was controlled without affecting transfection efficiency. This system revealed that (i) low expression of both mutants exerted toxicity sensitive to both Ac-DEVD-CHO (DEVD) and glutathione ethyl ester (GEE), whereas wild-type APP (wtAPP) only at higher expression levels caused GEE/DEVD-resistant death to lesser degrees; (ii) toxicity by the V642I mutation was entirely GEE/DEVD sensitive; and (iii) toxicity by higher expression of NL-APP was GEE/DEVD resistant. The GEE/DEVD-sensitive death was sensitive to pertussis toxin and was due to G(o)-interacting His(657)-Lys(676) domain. The GEE/DEVD-resistant death was due to C-terminal Met(677)-Asn(695). APP mutants lacking either domain unraveled elaborate intracellular cross-talk between these domains. EB18Q-APP, responsible for non-AD type of a human disease, only exerted GEE/DEVD-resistant death at higher expression. Therefore, (i) different FAD mutations in APP cause neuronal cell death through different cytoplasmic domains via different sets of mechanisms; (ii) expression levels of FAD genes are critical in activating specific death mechanisms; and (iii) toxicity by low expression of both mutants most likely reflects the pathogenetic mechanism of FAD.
  • T Niikura, N Murayama, Y Hashimoto, Y Ito, Y Yamagishi, M Matsuoka, Y Takeuchi, S Aiso, Nishimoto, I
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 274(2) 445-454 2000年8月  査読有り
    APP is a precursor of beta amyloid deposited in Alzheimer's disease (AD). Although genetic studies established that mutations in APP cause familial AD (FAD), the mechanism for neuronal death by FAD mutants has not been well understood. We established neuronal cells (F11/EcR/V642I cells) in which V642I APP was inducibly expressed by ecdysone. Treatment with ecdysone, but not vehicle, killed most cells within a few days, with rounding, shrinkage, and detachment as well as nuclear fragmentation. Death was suppressed by Ac-DEVD-CHO and pertussis toxin. Electron microscopic analysis revealed that apoptosis occurred in ecdysone-treated cells. V642I-APP-induced death was suppressed by the anti-AD factors estrogen and apoE2. These data demonstrate not only that expression of this FAD gene causes neuronal apoptosis, but that F11/EcR/V642I cells, the first neuronal cells with inducible FAD gene expression, provide a useful model system in investigating AD disorders. (C) 2000 Academic Press.
  • T Niikura, R Hirata, SC Weil
    BLOOD CELLS MOLECULES AND DISEASES 23(17) 337-349 1997年  査読有り
    The acute promyelocytic leukemia cell line, NB4, can be induced to differentiate to mature granulocytes by retinoic acid treatment. A novel retinoic acid-inducible cDNA clone, designated RI58, was isolated from a cDNA library constructed from retinoic acid-treated NB4 cells by differential hybridization. RI58 cDNA encodes a protein of 58kDa which has a similarity in its amino acids sequence to interferon (IFN)inducible proteins, In addition, RI58 was induced by recombinant human IFN-alpha (rhIFN-alpha) in NB4 cells, RI58 was detectable within 4 hours post-stimulation with rhIFN-alpha, while it took as long as 1 day after letinoic acid stimulation, Culture supernatant from retinoic acid-treated NB4 cells also induced RI58 expression similarly as rhIFN-alpha, This activity in culture supernatant was inhibited by anti-leukocyte IFN antiserum which showed specific reactivity to rhIFN-alpha. These results indicate that RI58 is induced by retinoic acid stimulation through autocrinally secreted IFN-alpha from NB4 cells. In the retinoic acid-treated NB4 cells, the expression of RI58 was increased along the process of differentiation. On the other hand, it was expressed constitutively in untreated non-hematopoietic cell lines and mature hematopoietic cell lines.
  • AK BASAK, DI STUART, T NIKURA, DHL BISHOP, DC KELLY, A FEARN, RW TITBALL
    JOURNAL OF MOLECULAR BIOLOGY 244(5) 648-650 1994年12月  査読有り
    Alpha-toxin of Clostridium perfringes, cloned in Escherichia coli, has been purified and crystallized from ammonium sulphate using the hanging drop vapour diffusion method at 20 degrees C. The crystals diffract to a minimum Bragg spacing of 2.7 Angstrom, belong to the space group R32 (with a = b = 153.3 Angstrom, c = 95.4 Angstrom, alpha = beta = 90 degrees and gamma = 120 degrees) and contain a single polypeptide chain in the crystallographic unit.

MISC

 29

講演・口頭発表等

 41

共同研究・競争的資金等の研究課題

 19

その他

 3
  • 2013年 - 2015年
    「ヒトの生物科学」のタイトルで「ヒト」をキーワードとした生物学・医学・工学・化学の広範な領域のトピックを最近の知見を含めて解説する講義をコーディネートしている。全学共通科目として多くの受講者が興味を持ちかつより深い理解ができるよう工夫している。
  • 2012年 - 2015年
    少人数の学部科目において発表会形式のプレゼンテーションを実施し、受講者がプレゼンテーション資料の作成から発表までの一連の作業を経験することで、その後の卒業研究などに自発的に取り組む重要性を理解できるよう工夫している。