大阪公立大学大学院 医学研究科 認知症病態学

研究業績

主な原著論文

  1. Umeda T, Shigemori K, Uekado R, Matsuda K, Tomiyama T.* Hawaiian native herb Mamaki prevents dementia by ameliorating neuropathology and repairing neurons in four different mouse models of neurodegenerative diseases. GeroScience. https://doi.org/10.1007/s11357-023-00950-y
  2. Hatanaka Y, Umeda T, Shigemori K, Takeuchi T, Nagai Y, Tomiyama T.* C9orf72 Hexanucleotide Repeat Expansion-Related Neuropathology Is Attenuated by Nasal Rifampicin in Mice. Biomedicines. 10(5), 1080, 2022.
  3. Shigemori K, Nomura S, Umeda T, Takeda S, Tomiyama T.* Peripheral Aβ acts as a negative modulator of insulin secretion. Proc.Natl.Acad.Sci.USA. 119(12), e2117723119, 2022.
  4. Umeda T, Uekado R, Shigemori K, Eguchi H, Tomiyama T.* Nasal Rifampicin Halts the Progression of Tauopathy by Inhibiting Tau Oligomer Propagation in Alzheimer Brain Extract-Injected Mice. Biomedicines. 10(2), 297, 2022.
  5. Umeda T, Sakai A, Shigemori K, Yokota A, Kumagai T, Tomiyama T.* Oligomer-targeting prevention of neurodegenerative dementia by intranasal rifampicin and resveratrol combination -a preclinical study in model mice. Frontiers in Neuroscience. 15, 763476, 2021.
  6. Umeda T, Hatanaka Y, Sakai A, Tomiyama T.* Nasal Rifampicin Improves Cognition in a Mouse Model of Dementia with Lewy Bodies by Reducing α-Synuclein Oligomers. International Journal of Molecular Sciences. 22(16), 8453, 2021.
  7. Umeda T, Tanaka A, Sakai A, Yamamoto A, Sakane T, Tomiyama T.* Intranasal rifampicin for Alzheimer's disease prevention. Alzheimer's Dementia: Transl. Res. Clin. Intervent. 4, 304-313, 2018.
  8. Umeda T, Kimura T, Yoshida K, Takao K, Fujita Y, Matsuyama S, Sakai A, Yamashita M, Yamashita Y, Ohnishi K, Suzuki M, Takuma H, Miyakawa T, Takashima A, Morita T, Mori H, Tomiyama T.* Mutation-induced loss of APP function causes GABAergic depletion in recessive familial Alzheimer’s disease: analysis of Osaka mutation-knockin mice. Acta Neuropathol. Commun. 5, 59, 2017.
  9. Umeda T., Ono K., Sakai A., Yamashita M., Mizuguchi M, Klein W.L., Yamada M., Mori H., Tomiyama T.* Rifampicin is a candidate preventive medicine against amyloid β and tau oligomers. Brain 139, 1568-1586, 2016.
  10. Umeda T., Ramser E.M., Yamashita M., Nakajima K., Mori H., Silverman M.A.*, Tomiyama T.* Intracellular amyloid β oligomers impair organelle transport and induce dendritic spine loss in primary neurons. Acta Neuropathol. Commun. 3, 51, 2015.
  11. Umeda T., Eguchi H., Kunori Y., Matsumoto Y., Taniguchi T., Mori H., Tomiyama T.* Passive immunotherapy of tauopathy targeting pSer413-tau: a pilot study in mice. Ann. Clin. Transl. Neurol. 2, 241-255, 2015.
  12. Umeda T., Maekawa S., Kimura T., Takashima A., Tomiyama T.*, Mori H. Neurofibrillary tangle formation by introducing wild-type human tau into APP transgenic mice. Acta Neuropathol. 127, 685-698, 2014.
  13. Nomura S., Umeda T., Tomiyama T.*, Mori H. The E693Δ (Osaka) mutation in amyloid precursor protein potentiates cholesterol-mediated intracellular amyloid β toxicity via its impaired cholesterol efflux. J. Neurosci. Res. 91, 1541-1550, 2013.
  14. Umeda T., Yamashita T., Kimura T., Ohnishi K., Takuma H., Ozeki T., Takashima A., Tomiyama T., Mori H.* Neurodegenerative disorder FTDP-17 related tau intron 10 +16C→T mutation increases tau exon 10 splicing and causes tauopathy in transgenic mice. Am. J. Pathol. 183, 211-225, 2013.
  15. Umeda T., Tomiyama T.*, Kitajima E., Idomoto T., Nomura S., Lambert M.P., Klein W.L., Mori H. Hypercholesterolemia accelerates intraneuronal accumulation of Aβ oligomers resulting in memory impairment in Alzheimer’s disease model mice. Life Sci. 91, 1169-1176, 2012.
  16. Umeda T., Tomiyama T.*, Sakama N., Tanaka S., Lambert M.P., Klein W.L., Mori H. Intraneuronal amyloid β oligomers cause cell death via endoplasmic reticulum stress, endosomal/lysosomal leakage, and mitochondrial dysfunction in vivo. J. Neurosci. Res. 89, 1031-1042, 2011.
  17. Tomiyama T.*, Matsuyama S., Iso H., Umeda T., Takuma H., Ohnishi K., Ishibashi K., Teraoka R., Sakama N., Yamashita T., Nishitsuji K., Ito K., Shimada H., Lambert M.P., Klein W.L., Mori H. A mouse model of amyloid β oligomers: Their contribution to synaptic alteration, abnormal tau phosphorylation, glial activation, and neuronal loss in vivo. J. Neurosci. 30, 4845-4856, 2010.
  18. Umeda T., Mori H., Zheng H., Tomiyama T.* Regulation of cholesterol efflux by amyloid β secretion. J. Neurosci. Res. 88, 1985-1994, 2010.
  19. Nishitsuji K., Tomiyama T.*, Ishibashi K., Ito K., Teraoka R., Lambert M.P., Klein W.L., Mori H. The E693Δ mutation in amyloid precursor protein increases intracellular accumulation of amyloid β oligomers and causes endoplasmic reticulum stress-induced apoptosis in cultured cells. Am. J. Pathol. 174, 957-969, 2009.
  20. Takuma H., Teraoka R., Mori H., Tomiyama T.* Amyloid β E22Δ variant induces synaptic alteration in mouse hippocampal slices. Neuroreport 19, 615-619, 2008.
  21. Tomiyama T., Nagata T., Shimada H., Teraoka R., Fukushima A., Kanemitsu H., Takuma H., Kuwano R., Imagawa M., Ataka S., Wada Y., Yoshioka E., Nishizaki T., Watanabe Y., Mori H.* A new amyloid β variant favoring oligomerization in Alzheimer's-type dementia. Ann. Neurol. 63, 377-387, 2008.
  22. Matsuyama S., Teraoka R., Mori H., Tomiyama T.* Inverse correlation between amyloid precursor protein and synaptic plasticity in transgenic mice. Neuroreport 18, 1083-1087, 2007.

最近の主な共著論文

    1. Amano A, Sanjo N, Araki W, Anraku Y, Nakaki M, Matsubara E, Tomiyama T, Nagata T, Tsumoto K, Kataoka K, Yokota T.* Peripheral administration of nanomicelle-encapsulated anti-Aβ oligomer fragment antibody reduces various toxic Aβ species in the brain. J Nanobiotechnology. 21, 36 ,2023. 
    2. Yanagisawa D, Ibrahim NF, Taguchi H, Morikawa S, Tomiyama T, Tooyama I.* Fluorine-19 Magnetic Resonance Imaging for Detection of Amyloid β Oligomers Using a Keto Form of Curcumin Derivative in a Mouse Model of Alzheimer’s Disease. Molecules. 26, 1362, 2021. 
    3. Yanagisawa D, Kato T, Taguchi H, Shirai N, Hirao K, Sogabe T, Tomiyama T, Gamo K, Hirahara Y, Kitada M, Tooyama I.* Keto form of curcumin derivatives strongly binds to Aβ oligomers but not fibrils. Biomaterials. 270, 120686, 2021. 
    4. Shimada H, Minatani S, Takeuchi J, Takeda A, Kawabe J, Wada Y, Mawatari A, Watanabe Y, Shimada H, Higuchi M, Suhara T, Tomiyama T, Itoh Y.* Heavy Tau Burden with Subtle Amyloid β Accumulation in the Cerebral Cortex and Cerebellum in a Case of Familial Alzheimer’s Disease with APP Osaka Mutation. Int. J. Mol. Sci. 21, 4443, 2020. 
    5. Kubo A, Ueda S, Yamane A, Wada-Kakuda S, Narita M, Matsuyama M, Nomori A, Takashima A, Kato T, Onodera O, Goto M, Ito M, Tomiyama T, Mori H, Murayama S, Ihara Y, Misonou H, Miyasaka T.* Ectopic Expression Induces Abnormal Somatodendritic Distribution of Tau in the Mouse Brain. J. Neurosci. 39, 6781-6797, 2019.
    6. Ohshima Y, Taguchi K, Mizuta I, Tanaka M, Tomiyama T, Kametani F, Yabe-Nishimura C, Mizuno T, Tokuda T.* Mutations in the β-amyloid precursor protein in familial Alzheimer's disease increase Aβ oligomer production in cellular models. Heliyon 4, e00511, 2018.
    7. Palavicini JP, Wang C, Chen L, Hosang K, Wang J, Tomiyama T, Mori H, Han X.* Oligomeric amyloid-beta induces MAPK-mediated activation of brain cytosolic and calcium-independent phospholipase A2 in a spatial-specific manner. Acta Neuropathol. Commun. 5, 56, 2017.
    8. Chambers JK, Tokuda T, Uchida K,* Ishii R, Tatebe H, Takahashi E, Tomiyama T,* Une Y, Nakayama H. The domestic cat as a natural animal model of Alzheimer's disease. Acta Neuropathol. Commun. 3, 78, 2015.

主な英文総説

  1. Tomiyama T.* & Shimada H. APP Osaka mutation in familial Alzheimer’s disease – Its discovery, phenotypes, and mechanism of recessive inheritance. Int. J. Mol. Sci. 21, 1413, 2020.

主な和文総説

  1. 富山貴美. 神経変性疾患を適応とする低分子化合物. 実験医学2023年増刊号「いま新薬で加速する神経変性疾患研究」, 41, 2004-2012, 2023.
  2. 富山貴美. Rifampicinのドラッグ・リポジショニングによる認知症予防薬の開発. 臨床精神薬理 26, 167-174, 2023.
  3. 富山貴美. リファンピシンの抗オリゴマー作用と認知症予防のための新たな投与法. In: 技術情報協会編. 認知症の早期診断技術と進行抑制/予防薬・機能性食品の開発. 技術情報協会, 東京, pp261-271, 2019.
  4. 富山貴美. アルツハイマー病の最新メカニズム、モデルマウスの作製と創薬への応用. In: 技術情報協会編. 動物/疾患モデルの作製技術・病態解析・評価手法. 技術情報協会, 東京, pp314-324, 2017.
  5. 富山貴美. タウ免疫療法 ─現状と展望. 実験医学2017年増刊号「認知症 発症前治療のために解明すべき分子病態は何か?」, 35, 2148-2154, 2017.
  6. 富山貴美. ドラッグ・リポジショニングによる抗認知症薬の探索. 実験医学2017年増刊号「認知症 発症前治療のために解明すべき分子病態は何か?」, 35, 2159-2165, 2017.
  7. 富山貴美. タウ免疫療法. 神経治療学 33, 428-432, 2016.
  8. 富山貴美. 新しいタウ抗体の開発と新たな認知症予防薬開発の試み. 生体の科学 67, 335-339, 2016.
  9. 富山貴美. AβオリゴマーとAPP Osaka変異. 医学のあゆみ 257, 389-394, 2016.
  10. 富山貴美. 期待されるアルツハイマー病の根治療法. Mebio 32, 6, 59-68, 2015.
  11. 富山貴美. タウ免疫療法の現況. Dementia Japan 29, 195-203, 2015.
  12. 富山貴美. 病因論に基づくADの診断・治療法の開発と変遷. BIO Clinica 29, 630-634, 2014.
  13. 富山貴美. アルツハイマー病の分子メカニズムを探る. からだの科学 278, 25-29, 2013.
  14. 富山貴美. Aβオリゴマーを標的とするアルツハイマー病治療薬の展望. Dementia Japan 26, 300-310, 2012.
  15. 富山貴美. 細胞内Aβオリゴマーによるアルツハイマー病発症機構. Dementia Japan 26, 225-241, 2012.
  16. 富山貴美. 認知症の分子遺伝学. 臨床検査, 56, 31-39, 2012.
  17. 富山貴美. AβオリゴマーとAβ凝集. 日本臨床, 69, 増刊号8「認知症学(上)」, 54-58, 2011.
  18. 富山貴美. 病因分子としてのβアミロイド. Brain and Nerve 62, 691-699, 2010.
  19. 富山貴美. Aβオリゴマー仮説―アルツハイマー病の新しい考え方―. Dementia Japan 24, 37-45, 2010.
  20. 富山貴美. βアミロイドオリゴマーとシナプス機能障害. Cognition and Dementia 8, 209-215, 2009.
  21. 富山貴美. 新しいAPP変異 (E693Δ) の同定とAβオリゴマー仮説. Dementia Japan 22, 26-35, 2008.