研究業績

原著論文

Ikeda M, Watanabe T, Ito A, Fujimuro M. Herpes simplex virus 1 infection induces ubiquitination of UBE1a. Biochem. J. 478, 261-279, 2021

Dileep KV, Sakai N, Ihara K, Kato-Murayama M, Nakata A, Ito A, Sivaraman DM, Shin JW, Yoshida M, Shirouzu M, Zhang KYJ. Piperidine-4-carboxamide as a new scaffold for designing secretory glutaminyl cyclase inhibitors. Int. J. Biol. Macromol. 170, 415-423, 2021

Ikeda M, Ito A, Sekine Y, Fujimuro M. UBE1a Suppresses Herpes Simplex Virus-1 Replication. Viruses 12, 1391, 2020

Shiroma Y, Fujita G, Yamamoto T, Takahashi RU, Kumar A, Zhang KYJ, Ito A, Osada H, Yoshida M, Tahara H. Identification of a Selective RelA Inhibitor Based on DSE-FRET Screening Methods. Int. J. Mol. Sci. 21, 9150, 2020

Maemoto Y, Maruyama T, Nemoto K, Baba T, Motohashi M, Ito A, Tagaya M, Tani K. DDHD1, but Not DDHD2, Suppresses neurite outgrowth in SH-SY5Y and PC12 cells by regulating protein transport from recycling endosomes. Front. Cell Dev. Biol. 8, 670, 2020.

Radwan MO, Ciftci HI, Ali TFS, Koga R, Tateishi H, Nakata A, Ito A, Yoshida M, Fujita M, Otsuka M. Structure activity study of S-trityl-cysteamine dimethylaminopyridine derivatives as SIRT2 inhibitors: Improvement of SIRT2 binding and inhibition. Bioorg. Med. Chem. Lett. 30, 127458, 2020

Matsumoto N, Miyamoto Y, Hattori K, Ito A, Harada H, Oizumi H, Ohbuchi K, Mizoguchi K, Yamauchi J. PP1C and PP2A are p70S6K Phosphatases Whose Inhibition Ameliorates HLD12-Associated Inhibition of Oligodendroglial Cell Morphological Differentiation. Biomedicines 8, 89, 2020

Fujikawa K, Nakahara K, Takasugi N, Nishiya T, Ito A, Uchida K, Uehara T. S-Nitrosylation at the active site decreases the ubiquitin-conjugating activity of ubiquitin-conjugating enzyme E2 D1 (UBE2D1), an ERAD-associated protein. Biochem. Biophys. Res. Commun. 524, 910-915, 2020.

Nagata A, I Fumiko, Sasho A, Sugita K, Suzuki R, Hinata H, Shimoda Y, Suzuki E, Maemoto Y, Inagawa T, Fujikawa Y, Ikeda E, Fujii C, Inoue H. The evolutionarily conserved deubiquitinase UBH1/UCH-L1 augments DAF7/TGF-β signaling, inhibits dauer larva formation, and enhances lung tumorigenesis. J Biol Chem. 295, 9105-9120 2020

Nakahara, K., Fujikawa, K., Hiraoka H., Miyazaki, I., Asanuma, M., Ito, A., Takasugi, N., Uehara, T. Attenuation of macrophage migration inhibitory factor-stimulated signaling via S-nitrosylation. Biol. Pharm. Bull. 42, 1044–1047, 2019

Radwan MO, Ciftci HI, Ali TFS, Ellakwa DE, Koga, R., Tateishi, H., Nakata, A., Ito, A., Yoshida, M., Okamoto, Y., Fujita, M., Otsuka, M. Antiproliferative S-Trityl-l-Cysteine -Derived Compounds as SIRT2 Inhibitors: Repurposing and Solubility Enhancement. Molecules. 24, pii: E3295, 2019

Nomura, Y., Thuaud F., Sekine, D., Ito, A., Maeda, S., Koshino, H., Hashizume, D., Muranaka, A., Cruchter T., Uchiyama, M., Ichikawa, S., Matsuda, A., Yoshida, M., Hirai, G., Sodeoka, M. Synthesis of All Stereoisomers of Monomeric Spectomycin A1/A2 and Evaluation of Their Protein SUMOylation-Inhibitory Activity. Chemistry 25, 8387-8392, 2019

Mitsui, E., Yoshida, S., Shinoda, Y., Matsumori, Y., Tsujii, H., Tsuchida, M., Wada, S., Hasegawa, M, Ito, A., Mino, K., Onuki, T., Yoshida, M., Sasaki, R., Mizukami, T. Identification of ryuvidine as a KDM5A inhibitor. Sci Rep. 9, 9952, 2019

Ali TFS, Ciftci HI, Radwan MO, Koga, R., Ohsugi, T., Okiyama, Y., Honma, T., Nakata, A., Ito, A., Yoshida, M., Fujita, M., Otsuka, M. New SIRT2 inhibitors: Histidine-based bleomycin spin-off. Bioorg. Med. Chem. 27, 1767-1775, 2019

Hayashi, Y., Harada, Y., Kagiyama, Y., Nishikawa, S., Ding, Y., Imagawa, J., Shingai, N., Kato, N., Kitaura, J., Hokaiwado, S., Maemoto, Y., Ito, A., Matsui, H., Kitabayashi, I., Iwama, A., Komatsu, N., Kitamura, T., Harada, H. NUP98-HBO1-fusion generates phenotypically and genetically relevant chronic myelomonocytic leukemia pathogenesis. Blood Adv. 3, 1047-1060, 2019

Hiranaka S, Tega Y, Higuchi K, Kurosawa T, Deguchi Y, Arata M, Ito A, Yoshida M, Nagaoka Y, Sumiyoshi T. Design, Synthesis, and Blood−Brain Barrier Transport Study of Pyrilamine Derivatives as Histone Deacetylase Inhibitors. ACS Med. Chem. Lett. 54, 9202-9205, 2018

Sohtome Y, Shimazu T, Barjau J, Fujishiro S, Akakabe M, Terayama N, Dodo K, Ito A, Yoshida M, Shinkai Y, Sodeoka M. Unveiling epidithiodiketopiperazine as a non-histone arginine methyltransferase inhibitor by chemical protein methylome analyses. Chem. Commun. 54, 9202-9205, 2018

Maruyama T, Baba T, Maemoto Y, Hara-Miyauchi C, Hasegawa-Ogawa M, Okano HJ, Enda Y, Matsumoto K, Arimitsu N, Nakao K, Hamamoto H6, Sekimizu K, Ohto-Nakanishi T, Nakanishi H, Tokuyama T, Yanagi S, Tagaya M, Tani K. Loss of DDHD2, whose mutation causes spastic paraplegia, promotes reactive oxygen speciesgeneration and apoptosis. Cell Death Dis. 9(8):797. 2018

Hirano T, Fujiwara T, Niwa H, Hirano M, Ohira K, Okazaki Y, Sato S, Umehara T, Maemoto Y, Ito A, Yoshida M, Kagechika H. Development of Novel Inhibitors for Histone Methyltransferase SET7/9 based on Cyproheptadine. ChemMedChem. 13(15):1530-1540. 2018

Nakamura F, Kudo K, Tomachi Y, Nakata A, Takemoto M, Ito A, Tabei H, Arai D, de Voogd N, Yoshida M, Nakao Y, Fusetani N, Halistanol Sulfates I and J, New SIRT1-3 Inhibitory Steroid Sulfates from a Marine Sponge of the Genus Halichondria. J. Antibiot. J Antibiot (Tokyo), 71(4):483 2018

Kuroki S, Baba S, Okashita N, Yano M, Yamaguchi M, Kitano S, Miyachi H, Ito A, Yoshida M, Tachibana M. Rescuing the aberrant sex development of H3K9 demethylase Jmjd1a-deficient mice by modulating H3K9 methylation balance. PLoS Genet. 13(9):e1007034, 2017

#Kudo N, #Ito A, Arata M, Nakata A, Yoshida M. Identification of a novel small molecule that inhibits deacetylase but not defattyacylase reaction catalyzed by SIRT2. Philos. Trans. B. Philos Trans R Soc Lond B Biol Sci. 373(1748), 2018

Ueda M, Matsui A, Tanaka M, Nakamura T, Abe T, Sako K, Sasaki T, Kim JM, Ito A, Nishino N, Shimada H, Yoshida M, Seki M. The Distinct Roles of Class I and II RPD3-Like Histone Deacetylases in Salinity Stress Response. Plant Physiol. 175(4):1760-1773, 2017

Okochi-Takada E, Hattori N, Ito A, Niwa T, Wakabayashi M, Kimura K, Yoshida M, Ushijima T. Establishment of a high-throughput detection system for DNA demethylating agents. Epigenetics. 13(2):147-155, 2018

Noritsugu K, *Ito A, Nakao Y, Yoshida M. Identification of zinc finger transcription factor EGR2 as a novel acetylated protein. Biochem. Biophys. Res. Commun. 489, 455-459, 2017

Kurita K, Sakamoto T, Yagi N, Sakamoto Y, Ito A, Nishino N, Sako K, Yoshida M, Kimura H, Seki M, Matsunaga S. Live imaging of H3K9 acetylation in plant cells. Sci. Rep. 7, 45894, 2017

Sunami Y, Araki M, Kan S, Ito A, Hironaka Y, Imai M, Morishita S, Ohsaka A, Komatsu N. Histone acetyltransferase p300/CREB-binding protein-associated factor (PCAF) is required for all-trans-retinoic acid-induced granulocytic differentiation inleukemia cells. J. Biol. Chem. 292, 2815-2829, 2017

Narita K, Katoh Y, Ojima K, Dan S, Yamori T, Ito A, Yoshida M, Katoh T. Total Synthesis of the Depsipeptide FR901375 and Preliminary Evaluation of Its Biological Activity. Eur. J. Org. Chem. 121, 5667–5677, 2016

Ueoka R, Hitora Y, Ito A, Yoshida M, Okada S, Takada K, Matsunaga S. Curacin E from the Brittle Star Ophiocoma scolopendrina. J. Nat. Prod. 79, 2754-2757, 2016

Fukuda I, Hirohama M, *Ito A, Tariq M, Igarashi Y, Saitoh H, Yoshida M. Inhibition of protein SUMOylation by natural quinones. J. Antibiot. 269, 776-779, 2016

Shah AA, *Ito A, Nakata A, Yoshida M. Identification of a selective SIRT2 inhibitor and its anti-breast cancer activity. Biol. Pharm. Bull. 39, 1739-1742, 2016

Takada K, Imae Y, Ise Y, Ohtsuka S, Ito A, Okada S, Yoshida M, Matsunaga S. Yakushinamides, Polyoxygenated Fatty Acid Amides That Inhibit HDACs and SIRTs, from the Marine Sponge Theonella swinhoei. J. Nat. Prod. 79, 2384-2390, 2016

Fujiwara T, Ohira K, Urushibara K, Ito A, Yoshida M, Kanai M, Tanatani A, Kagechika H, Hirano T. Steric structure-activity relationship of cyproheptadine derivatives as inhibitors of histone methyltransferase Set7/9. Bioorg. Med. Chem. 24, 4318-4323, 2016

Matsumoto Y, Ito A, Uesugi M, Kittaka A. Efficient N-Acyldopamine Synthesis. Chem. Pharm. Bull. 64, 935-940, 2016

Narita K, Matsuhara K, Itoh J, Akiyama Y, Dan S, Yamori T, Ito A, Yoshida M, Katoh T. Synthesis and biological evaluation of novel FK228 analogues as potential isoform selective HDAC inhibitors. Eur. J. Med. Chem. 121, 592-609, 2016

Takemoto Y, *Ito A, Niwa H, Okamura M, Fujiwara T, Hirano T, Handa N, Umehara T, Sonoda T, Ogawa K, Tariq M, Nishino N, Dan S, Kagechika H, Yamori T, Yokoyama S, Yoshida M. Identification of Cyproheptadine as an Inhibitor of SET Domain Containing Lysine Methyltransferase 7/9 (Set7/9) That Regulates Estrogen-Dependent Transcription. J. Med. Chem. 59, 3650-3660, 2016

Nakata S, Watanabe T, Nakagawa K, Takeda H, Ito A, Fujimuro M. The dynamics of histone H2A ubiquitination in HeLa cells exposed to rapamycin, ethanol, hydroxyurea, ER stress, heat shock and DNA damage. Biochem. Biophys. Res. Commun. 472, 46-52, 2016

Kumar A, Ito A, Hirohama M, Yoshida M, Zhang KY. Identification of new SUMO activating enzyme 1 inhibitors using virtual screening and scaffold hopping. Bioorg. Med. Chem. Lett. 26, 1218-1223, 2016

Tariq M, *Ito A, Ishfaq M, Bradshaw E, Yoshida M. Eukaryotic translation initiation factor 5A (eIF5A) is essential for HIF-1α activation in hypoxia. Biochem. Biophys. Res. Commun. 470, 417-424, 2016

Nakaoka S, Sasaki K, Ito A, Nakao Y, Yoshida M. A Genetically Encoded FRET Probe to Detect Intranucleosomal Histone H3K9 or H3K14 Acetylation Using BRD4, a BET Family Member. ACS Chem. Biol. 11, 729-733, 2016

Ito A, Shimazu T, Maeda S, Shah AA, Tsunoda T, Iemura S, Natsume T, Suzuki T, Motohashi H, Yamamoto M, Yoshida M. The subcellular localization and activity of cortactin is regulated by acetylation and interaction with Keap1. Sci. Signal. 8, ra120, 2015

Okuda K, Ito A, Uehara T. Regulation of Histone Deacetylase 6 Activity via S-Nitrosylation. Biol. Pharm. Bull. 38, 1434-1437, 2015

Feldman JL, Dittenhafer-Reed KE, Kudo N, Thelen JN, Ito A, Yoshida M, Denu JM. Kinetic and Structural Basis for Acyl-Group Selectivity and NAD(+) Dependence in Sirtuin-Catalyzed Deacylation. Biochemistry. 54, 3037-3050, 2015

Islam MS, Islam MN, Hoque MA, Nishino N, Kato T, Kim HJ, Ito A, Yoshida M. An efficient synthesis of SK-658 and its analogs as potent histone deacetylase inhibitors. Bioorg. Chem. 59, 145-150, 2015

Bai D, Nakao R, Ito A, Uematsu H, Senpuku H. Immunoreactive antigens recognized in serum samples from mice intranasally immunized with Porphyromonas gingivalis outer membrane vesicles. Pathog. Dis. 73, pii: ftu006., 2015 Le Bescont A, Vitte AL, Debernardi A, Curtet S, Buchou T, Vayr J, de Reyniès A, Ito A, Guardiola P, Brambilla C, Yoshida M, Brambilla E, Rousseaux S, Khochbin S. Receptor-Independent Ectopic Activity of Prolactin Predicts Aggressive Lung Tumors and Indicates HDACi-Based Therapeutic Strategies. Antioxid. Redox Signal. 23, 1-14, 2015

Maemoto Y, Ono Y, Kiso S, Shibata H, Takahara T, Sorimachi H, Maki M. Involvement of calpain-7 in epidermal growth factor receptor degradation via the endosomal sorting pathway. FEBS Journal. 281, 3642-3655, 2014

#Voet A, #Ito A, Hirohama M, Matsuoka S, Tochio N, Kigawa T, Yoshida M, Zhang KY. Discovery of Small Molecule Inhibitors Targeting the SUMO–SIM Interaction Using a Protein Interface Consensus Approach. MedChemComm. 5, 783-786, 2014 #These authors contributed equally

Islam MN, Islam MS, Hoque MA, Kato T, Nishino N, Ito A, Yoshida M. Bicyclic tetrapeptide histone deacetylase inhibitors with methoxymethyl ketone and boronic acid zinc-binding groups. Bioorg. Chem. 57, 121-126, 2014

#Kumar A, #Ito A, Hirohama M, Yoshida M, Zhang KY. Identification of sumoylation inhibitors targeting a predicted pocket in ubc9. J. Chem. Inf. Model. 54, 784-793, 2014 #These authors contributed equally 

Hoque MA, Islam MS, Islam MN, Kato T, Nishino N, Ito A, Yoshida M. Design and synthesis of mono and bicyclic tetrapeptides thioester as potent inhibitor of histone deacetylases. Amino Acids. 46, 2435-2444, 2014

Islam MN, Islam MS, Hoque MA, Kato T, Nishino N, Ito A, Yoshida M. Bicyclic tetrapeptides as potent HDAC inhibitors: Effect of aliphatic loop position and hydrophobicity on inhibitory activity. Bioorg. Med. Chem. 22, 3862-3870, 2014

Fukui Y, Narita K, Dan S, Yamori T, Ito A, Yoshida M, Katoh T. Total synthesis of burkholdacs A and B and 5,6,20-tri-epi-burkholdac A: HDAC inhibition and antiproliferative activity. Eur. J. Med. Chem. 76, 301-313, 2014

Khan K, Schneider-Poetsch T, Ishfaq M, Ito A, Yoshimoto R, Mukaida N, Yoshida M. Splicing inhibition induces gene expression through canonical NF-κB pathway and extracellular signal-related kinase activation. FEBS Lett. 588, 1053-1057, 2014

#Kumar A, #Ito A, Takemoto M, Yoshida M, Zhang KY. Identification of 1,2,5-oxadiazoles as a new class of SENP2 inhibitors using structure based virtual screening. J. Chem. Inf. Model. 54, 870-880, 2014 #These authors contributed equally 

Chi H, Takemoto Y, Nsiama TK, Kato T, Nishino N, Ito A, Yoshida M. Design and synthesis of peptide-MCA substrates for a novel assay of histone methyltransferases and their inhibitors. Bioorg. Med. Chem. 22, 268-275, 2014

Takemoto M, Kawamura Y, Hirohama M, Yamaguchi Y, Handa H, Saitoh H, Nakao Y, Kawada M, Khalid K, Koshino H, Kimura K, *Ito A, Yoshida M. Inhibition of protein SUMOylation by davidiin, an ellagitannin from Davidia involucrata. J. Antibiot. 67, 335-338, 2014

Hirohama M, Voet AR, Ozawa T, Saitoh H, Nakao Y, Zhang KY, *Ito A, Yoshida M. Assay methods for small ubiquitin-like modifier (SUMO)-SUMO-interacting motif (SIM) interactions in vivo and in vitro using a split-luciferase complementation system. Anal. Biochem. 448, 92-94, 2014

Maemoto Y, Kiso S, Shibata H, Maki M. Analysis of limited proteolytic activity of calpain-7 using non-physiological substrates in mammalian cells. FEBS Journal. 280 2594-2607, 2013

Maemoto Y, Shibata H, Maki M. Determination of Phosphorylation Site in CHMP1A. Bioscience, Biotechnology, and Biochemistry, 77, 1317-1319, 2013

Hirohama M, Kumar A, Fukuda I, Matsuoka S, Igarashi Y, Saitoh H, Takagi M, Shin-ya K, Honda K, Kondoh Y, Saito T, Nakao Y, Osada H, Zhang KY, Yoshida M, *Ito A. Spectomycin B1 as a novel SUMOylation inhibitor that directly binds to SUMO E2. ACS Chem. Biol. 8, 2635-2642, 2013

Hao R, Nanduri P, Rao Y, Panichelli RS, Ito A, Yoshida M, Yao TP. Proteasomes activate aggresome disassembly and clearance by producing unanchored ubiquitin chains. Mol. Cell 51, 819-828, 2013

Kumar A, Ito A, Hirohama M, Yoshida M, Zhang KY. Identification of quinazolinyloxy biaryl urea as a new class of SUMO activating enzyme 1 inhibitors. Bioorg. Med. Chem. Lett. 23, 5145-5149, 2013

Arai T, Ashraful Hoque M, Nishino N, Kim HJ, Ito A, Yoshida M. Cyclic tetrapeptides with -SS- bridging between amino acid side chains for potent histone deacetylases’ inhibition. Amino Acids. 45, 835-843, 2013

Matsubara K, Lee AR, Kishigami S, Ito A, Matsumoto K, Chi H, Nishino N, Yoshida M, Hosoi Y. Dynamics and regulation of lysine-acetylation during one-cell stage mouse embryos. Biochem. Biophys. Res. Commun. 434, 1-7, 2013

Kumar A, Ito A, Hirohama M, Yoshida M, Zhang KY. Identification of sumoylation activating enzyme 1 inhibitors by structure-based virtual screening. J. Chem. Inf. Model. 53, 809-820, 2013

Fujishiro S, Dodo K, Iwasa E, Teng Y, Sohtome Y, Hamashima Y, Ito A, Yoshida M, Sodeoka M. Epidithiodiketopiperazine as a pharmacophore for protein lysine methyltransferase G9a inhibitors: Reducing cytotoxicity by structural simplification. Bioorg. Med. Chem. Lett. 23, 733-736, 2013

Narita K, Fukui Y, Sano Y, Yamori T, Ito A, Yoshida M, Katoh T. Total synthesis of bicyclic depsipeptides spiruchostatins C and D and investigation of their histone deacetylase inhibitory and antiproliferative activities. Eur. J. Med. Chem. 60, 295-304, 2012

Ishfaq M, Maeta K, Maeda S, Natsume T, *Ito A, Yoshida M. The Role of Acetylation in the Subcellular Localization of an Oncogenic Isoform of Translation Factor eIF5A. Biosci. Biotechnol. Biochem. 76, 2165-2167, 2012

Hoque MA, Arai T, Nishino N, Kim HJ, Ito A, Yoshida M. Cyclic tetrapeptides with thioacetate tails or intramolecular disulfide bridge as potent inhibitors of histone deacetylases. Bioorg. Med. Chem. Lett. 22, 6770-6772, 2012

Cho HS, Shimazu T, Toyokawa G, Daigo Y, Maehara Y, Hayami S, Ito A, Masuda K, Ikawa N, Field HI, Tsuchiya E, Ohnuma S, Ponder BA, Yoshida M, Nakamura Y, Hamamoto R. Enhanced HSP70 lysine methylation promotes proliferation of cancer cells through activation of Aurora kinase B. Nat. Commun. 3, 1072, 2012

Ishfaq M, Maeta K, Maeda S, Natsume T, *Ito A, Yoshida M. Acetylation regulates subcellular localization of eukaryotic translation initiation factor 5A (eIF5A). FEBS Lett. 586, 3236-3241, 2012

Yoshihara H, Fukushima T, Hakuno F, Saeki Y, Tanaka K, Ito A, Yoshida M, Iemura S, Natsume T, Asano T, Chida K, Girnita L, Takahashi S. Insulin/insulin-like growth factor (IGF) stimulation abrogates an association between a deubiquitinating enzyme USP7 and insulin receptor substrates (IRSs) followed by proteasomal degradation of IRSs. Biochem. Biophys. Res. Commun. 423, 122-127, 2012

Kamemura K, Ogawa M, Ohkubo S, Ohtsuka Y, Shitara Y, Komiya T, Maeda S, Ito A, Yoshida M. Depression of mitochondrial metabolism by downregulation of cytoplasmic deacetylase, HDAC6. FEBS Lett. 586, 1379-1383, 2012

Matsushita K, Kajiwara T, Tamura M, Satoh M, Tanaka N, Tomonaga T, Matsubara H, Shimada H, Yoshimoto R, Ito A, Kubo S, Natsume T, Levens D, Yoshida M, Nomura F. SAP155-mediated splicing of FUSE-binding protein-interacting repressor (FIR) serves as a molecular switch for c-myc gene expression. Mol. Cancer Res. 10, 787-799, 2012

Kobayashi H, Harada H, Nakamura M, Futamura Y, Ito A, Yoshida M, Iemura SI, Shin-Ya K, Doi T, Takahashi T, Natsume T, Imoto M, Sakakibara Y. Comprehensive predictions of target proteins based on protein-chemical interaction using virtual screening and experimental verifications. BMC Chem. Biol. 12, 2, 2012

Takahashi M, Takemoto Y, Shimazu T, Kawasaki H, Tachibana M, Shinkai Y, Takagi M, Shin-Ya K, Igarashi Y, *Ito A, Yoshida M. Inhibition of histone H3K9 methyltransferases by gliotoxin and related epipolythiodioxopiperazines. J. Antibiot. 65, 263-265, 2012

Maemoto Y, Osako Y, Goto E, Nozawa E, Shibata H, Maki M. Calpain-7 binds to CHMP1B at its second α-helical region and forms a ternary complex with IST1. Journal of Biochemistry, 150, 411-421, 2011

Fukushima T, Okajima H, Yamanaka D, Ariga M, Nagata S, Ito A, Yoshida M, Asano T, Chida K, Hakuno F, Takahashi S. HSP90 interacting with IRS-2 is involved in cAMP-dependent potentiation of IGF-I signals in FRTL-5 cells. Mol. Cell Endocrinol. 344, 81-89, 2011

Islam MS, Bhuiyan MP, Islam MN, Nsiama TK, Oishi N, Kato T, Nishino N, Ito A, Yoshida M. Evaluation of functional groups on amino acids in cyclic tetrapeptides in histone deacetylase inhibition. Amino Acids 42, 2103-2120, 2011

Ito T, Umehara T, Sasaki K, Nakamura Y, Nishino N, Terada T, Shirouzu M, Padmanabhan B, Yokoyama S, *Ito A, *Yoshida M. Real-Time Imaging of Histone H4K12-Specific Acetylation Determines the Modes of Action of Histone Deacetylase and Bromodomain Inhibitors. Chem. Biol. 18, 495-507, 2011

Furumai R, Ito A, Ogawa K, Maeda S, SaIto A., Nishino N, Horinouchi S, Yoshida M. Histone deacetylase inhibitors block nuclear factor-κB-dependent transcription by interfering with RNA polymerase II recruitment. Cancer Sci. 102, 1081-1087, 2011

Oiso H, Furukawa N, Suefuji M, Shimoda S, Ito A, Furumai R, Nakagawa J, Yoshida M, Nishino N, Araki E. The role of class I histone deacetylase (HDAC) on gluconeogenesis in liver. Biochem. Biophys. Res. Commun. 404, 166-172, 2011

Osako Y, Maemoto Y, TanakaR, Suzuki H, Shibata H, Maki M. Autolytic activity of human calpain-7 is enhanced by ESCRT-III-related protein IST1 through MIT-MIM interaction. FEBS Journal, 227, 4412-4426, 2010

Iwasa E, Hamashima Y, Fujishiro S, Higuchi E, Ito A, Yoshida M, Sodeoka M. Total synthesis of (+)-chaetocin and its analogues: their histone methyltransferase G9a inhibitory activity. J. Am. Chem. Soc. 132, 4078-4079, 2010

Islam NM, Kato T, Nishino N, Kim HJ, Ito A, Yoshida M. Bicyclic peptides as potent inhibitors of histone deacetylases: optimization of alkyl loop length. Bioorg. Med. Chem. Lett. 20, 997-999, 2010

Fukuda I, *Ito A, Uramoto M, Saitoh H, Kawasaki H, Osad, H, *Yoshida M. Kerriamycin B inhibits protein SUMOylation. J. Antibiot. 62, 221-224, 2009

Fukuda I, *Ito A, Hirai G, Nishimura S, Kawasaki H, Saitoh H, Kimura K, Sodeoka M, Yoshida M. Ginkgolic acid inhibits protein SUMOylation by blocking formation of the E1-SUMO intermediate. Chem. Biol. 16, 133-140, 2009

Ueoka R, Ito A, Izumikawa M, Maeda S, Takagi M, Shin-ya K, Yoshida M, van Soest RW, Matsunaga S. Isolation of azaspiracid-2 from a marine sponge Echinoclathria sp. as a potent cytotoxin. Toxicon 53, 680-684, 2009

Dohi Y, Ikura T, Hoshikawa Y, Katoh Y, Ota K, Nakanome A, Muto A, Omura S, Ohta T, Ito A, Yoshida M, Noda T, Igarashi K. Bach1 inhibits oxidative stress-induced cellular senescence by impeding p53 function on chromatin. Nat. Struct. Mol. Biol. 15, 1246-1254, 2008

Kamemura K, *Ito A, Shimazu T, Matsuyama A, Maeda S, Yao TP, Horinouchi S, Khochbin S, Yoshida M. Effects of downregulated HDAC6 expression on the proliferation of lung cancer cells. Biochem. Biophys. Res. Commun. 374, 84-89, 2008

Kawaguchi Y, Ito A, Appella E, Yao TP. Charge modification at multiple C-terminal lysine residues regulates p53 oligomerization and its nucleus-cytoplasm trafficking. J. Biol. Chem. 281, 1394-1400, 2006

Saji S, Kawakami M, Hayashi S, Yoshida N, Hirose M, Horiguchi S, Itoh A, Funata N, Schreiber SL, Yoshida M, Toi M. Significance of HDAC6 regulation via estrogen signaling for cell motility and prognosis in estrogen receptor-positive breast cancer. Oncogene, 24, 4531-4539, 2005

Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao TP. The Deacetylase HDAC6 Regulates Aggresome Formation and Cell Viability in Response to Misfolded Protein Stress. Cell 115, 727-738, 2003

Ito A, Kawaguchi Y, Lai CH, Kovacs JJ, Higashimoto Y, Appella E, Yao TP. MDM2-HDAC1-mediated deacetylation of p53 is required for its degradation. EMBO J. 21, 6236-6345, 2002

Hubbert C, Guardiola A, Shao R, Kawaguchi Y, Ito A, Nixon A, Yoshida, M, Wang XF, Yao TP. HDAC6 is a microtubule-associated deacetylase. Nature 417, 455-458, 2002

Zhao X, Ito A, Kane CD, Liao TS, Bolger TA, Lemrow SM, Means AR, Yao TP. The Modular Nature of Histone Deacetylase HDAC4 Confers Phosphorylation-dependent Intracellular Trafficking. J. Biol. Chem. 276, 35042-35048, 2001

Ito A, Lai CH, Zhao X, Saito S, Hamilton MH, Appella E, Yao TP. p300/CBP-mediated p53 acetylation is commonly induced by p53 activating agents and inhibited by MDM2. EMBO J. 20, 1331-1340, 2001

Ito A, Uehara T, Nomura Y. Isolation of Ich-1S (caspase-2S)-binding protein that partially inhibits caspase activity. FEBS Lett. 470, 360-364, 2000

Ito A, Uehara T, Tokumitsu A, Okuma Y, Nomura Y. Possible involvement of cytochrome c release and sequential activation of caspases in ceramide-induced apoptosis in SK-N-MC cells. Biochim. Biophys. Acta. 1452, 263-274, 1999

Itano Y, Ito A, Uehara T, Nomura Y. Regulation of Bcl-2 protein expression in human neuroblastoma SH-SY5Y cells: positive and negative effects of protein kinases C and A, respectively. J. Neurochem. 67, 131-137, 1996

Okada F, Ito A, Hirokawa T, Tokumitsu Y Nomura Y. Long-term neuroleptic treatments counteract dopamine D2 agonist inhibition of adenylate cyclase but do not affect pertussis toxin ADP-ribosylation in the rat brain. Neurochem. Int. 28, 161-168, 1996

Okada F, Takahasi N, Ito A, Tokumitsu Y, Nomura Y. Acceleration of desipramine-induced changes on the dopamine receptor-coupled adenylate cyclase system by pertussis toxin. J. Neural Transm. Gen. Sect. 98, 133-142, 1994

Review Articles, Conference Presentations

総説

Yoshida M, Kudo N, Kosono S, Ito A. Chemical and structural biology of protein lysine deacetylases. Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 93, 297-321, 2017

Sasaki K, *Ito A, Yoshida M. Development of live-cell imaging probes for monitoring histone modifications. Bioorg. Med. Chem. 20, 1887-1892, 2012

Maki M, Maemoto Y, Osako Y, Shibata H. Evolutionary and physical linkage between calpains and penta-EF-hand Ca2+-binding proteins. FEBS Journal, 279, 1414-1421, 2012

学会発表

手賀悠真、平中誠弥、樋口慧、出口芳春、伊藤昭博、吉田稔、長岡康夫、住吉孝明、「H+/有機カチオン交換輸送体を標的としたHDAC阻害剤の脳へのデリバリー」、第39回生体膜と薬物の相互作用シンポジウム、2017年10月、金沢

平野智也、平野道丈、藤原敬士、梅原崇史、伊藤昭博、吉田稔、影近弘之、「特異な構造を有するCyproheptadine誘導体のSet7/9阻害剤としての構造展開」、第35回メディシナルケミストリーシンポジウム、2017年10月、名古屋

伊藤昭博、「NAD依存的脱アセチル化酵素SIRT2を標的とした創薬研究から見出された脂質代謝物の新しい機能の発見」、生体機能と創薬シンポジウム2017、2017年8月、京都 招待講演

Yoshida M and Ito A, “Chemical biology on deacetylation and deacylation by sirtuins”, FASEB Science research conferences “Reversible acetylation in health and disease”, August 2017, Big Sky, USA

Ito A, Noritsugu K, Suzuki T, Dohmae N, Yoshida M, “Long chain fatty acyl lysine modification of TEAD transcription factors”, FASEB Science research conferences “Reversible acetylation in health and disease”, August 2017, Big Sky, USA

伊藤昭博、室井誠、長田裕之、吉田稔、「翻訳因子eIF5AによるHIF-1αの活性制御」、第21回日本がん分子標的治療学会学術集会、2017年6月、福岡

伊藤昭博、横山翔、波多野稔子、松岡聖二、新真由美、Ashutosh Kumar、Kam Y.J. Zhang、浜本牧子、吉田稔、「ヒストンH3の27番目のリジン残基のメチル化を認識するCBX2タンパク質を標的とした化合物スクリーニング」、日本ケミカルバイオロジー学会第12回年会、2017年6月、札幌

伊藤昭博、「合成および内因性小分子化合物によるNAD依存的リジン脱アセチル化酵素の活性制御」、第11回日本エピジェネティクス研究会年会、2017年5月、東京 招待講演

伊藤昭博、横山翔、波多野稔子、松岡聖二、Ashutosh Kumar、Kam Y. J. Zhang、浜本牧子、吉田稔、「クロモドメインタンパク質CBX2を標的とした新しいがん治療法の開発」、日本薬学会第137年会、2017年3月、仙台

伊藤昭博、芹澤茉里子、Asad Ali Shah、前田里子、中田明子、三野光識、松岡聖二、松本洋太郎、有田誠、橘高敦史、上杉志成、吉田稔、「造腫瘍性代謝物様の活性を有する新規内因性脂質代謝物の発見」、第90回日本薬理学会年会、2017年3月、長崎

Ito A, “Discovery of novel lipid metabolites that may function as novel oncometabolites”, RIKEN Epigenetics in Tsukuba Recent advances in Epigenetic Engineering, Feburary 2017, Tukuba, Japan

Ito A, “Regulation of a NAD-dependent lysine deacetylase, SIRT2, by exogenous and endogenous small molecules”, INTERNATIONAL SEMINAR "Insulin-like Signaling and Nutrient Signaling: universal signaling for extension of healthy lifespan and improvement of quality for humans and animals, January 2017, Tokyo, Japan

伊藤昭博、「クロモドメインタンパク質CBX2を標的としたがん治療法の開発研究」、Promega Dynamic Connection 2016、2016年12月、横浜 招待講演

Ito A, Serizawa M, Shah AA, Mino K, Matsuoka S, Nakata A, Maeda S, Matsumoto Y, Nakao Y, Arita M, Kittaka A, Uesugi M, Yoshida M、“Discovery of novel lipid metabolites that possess oncometabolite-like properties”、第39回日本分子生物学会年会、2016年11-12月、横浜

Ito A, Takemoto Y, Fujiwara T, Hirano T, Niwa H, Handa N, Umehara T, Kagechika H, Yokoyama S, Yoshida M, “Identification of cyproheptadine as an inhibitor of lysine methyltransferase Set7/9 that regulates estrogen-dependent transcription”, International Symposium on Biomedical Engineering, November 2016, Tokyo, Japan

Ito A, Kudo N, Nakata A, Maeda S, Yoshida M,“Identification of a Selective SIRT2 Inhibitor and Its Antitumor Activit”, The 52nd International Conference on Medicinal Chemistry, July 2016, Caen, France

伊藤昭博、「NAD依存性脱アセチル化酵素SIRT2による細胞運動制御機構とその内因性阻害物質」、東京大学セミナー、2016年7月、東京

伊藤昭博、吉田稔、「NAD+依存的リジン脱アセチル化酵素SIRT2を標的としたがん治療法の開発」、第20回日本がん分子標的治療学会学術集会、2016年5月、別府

Ito A, Kudo N, Nakata A, Maeda S, Yoshida M, “Novel functions of fatty acid metabolites”, RIKEN-Max Planck Joint Research Center for Systems Chemical Biology 5th Annual Symposium, April 2016, Berlin, Germany

Ito A, Kudo N, Nakata A, Maeda S, Yoshida M, “Identification and crystallographic analysis of a small molecule selective SIRT2 inhibitor, and its antitumor activity”, 第89回日本薬理学会年会、2016年3月、横浜

伊藤昭博、「ヒストン修飾酵素を標的とした抗がん剤開」、理研シンポジウム 第3回創薬ワークショップ、2016年3月、横浜 招待講演

Ito A, “Identification of a small molecule selective SIRT2 inhibitor and its potential for an anticancer drug development”, The Kick-off Symposium in NCU, March 2016, Nagoya, Japan

Ito A, Kudo N, Nakata A, Maeda S, Yoshida M, “Identification of a small molecule selective SIRT2 inhibitor and its unique inhibitory mechanism”, International Symposium for RIKEN Epigenetics, February 2016, Wako, Japan

伊藤昭博、「SUMO化経路標的とした化合物の探索」、第13回SUMO研究会、2016年1月、大阪 招待講演

伊藤昭博、「タンパク質アセチル化による細胞運動制御〜基礎から応用さらに基礎研究へ〜」、関西学院大学セミナー、2016年1月、大阪 招待講演

Ito A, Kudo N, Nakata A, Maeda S, Yoshida M, “Novel functions of lipid metabolites as endogenous SIRT2 inhibitors”, The 8th Japan-Korea Chemical Biology Symposium, January 2016, Naha, Japan

伊藤昭博、「SUMO化経路標的とした化合物の探索」、第13回SUMO研究会、2016年1月、大阪

Ito A, Kudo N, Nakata A, Maeda S, Yoshida M, “Identification and crystallographic analysis of a small molecule selective SIRT2 inhibitor”, The International Chemical Congress of Pacific Basin Societies 2015, December 2015, Honolulu, USA

伊藤昭博、吉田稔、「NAD依存性脱アセチル化酵素SIRT2による細胞運動制御機構とその内因性阻害物質」、第2回北陸エピジェネティクス研究会、2015年11月、富山 招待講演

伊藤昭博、吉田稔、「Essential roles of eIF5A in HIF-1 stability and cancer cell survival in hypoxia」、第74回日本癌学会学術総会、2015年10月、名古屋

Ito A, Kudo N, Nakata A, Maeda S, Yoshida M, “Identification of a small molecule compound that selectively inhibits the NAD-dependent deacetylase SIRT2 by inducing conformational changes in the active site”, The 40th Naito Conference on Epigenetics—From Histone Code to Therapeutic Strategy, September 2015, Sapporo, Japan

伊藤昭博、吉田稔、「選択的SIRT2阻害剤の同定と立体構造解析」、第19回日本がん分子標的治療学会学術集会、2015年6月、松山

伊藤昭博、前田里子、吉田稔、「NAD依存的脱アセチル化酵素SIRT2による細胞運動制御機構」、日本薬学会第135年会、2015年3月、神戸 招待講演

Ito A, Yoshida M,“Identification of novel SENP1 inhibitors and their potential anti-tumor activities,” 第88回日本薬理学会年会、2015年3月、名古屋

伊藤昭博、「タンパク質アセチル化による細胞運動性の制御」、京都薬科大学特別研究セミナー、2015年3月、京都 招待講演

伊藤昭博、「タンパク質SUMO化を標的とした阻害剤の探索—タンパク質SUMO化酵素阻害剤およびSUMO-SIM結合阻害剤について—」、第12回SUMO研究会、2015年1月、大阪

Ito A, Yoshida M, “Identification of small molecule compounds that target protein SUMOylation and their potential anti-tumor activities”, 第73回日本癌学会学術総会、2014年9月、横浜 招待講演

伊藤昭博、吉田稔、「乳がん細胞におけるメチル化リジン結合タンパク質CBX2の機能解析」、第18回日本がん分子標的治療学会学術集会、2014年6月、仙台

伊藤昭博、波多野稔子、Muhammad Muddassar、Kam YJ Zhan、吉田稔、「乳がん細胞の増殖におけるメチル化リジン結合タンパク質CBX2の重要性の解明」、化学療法基盤支援活動第3回シンポジウム、2014年5月、名護

Ito A, Maeda S, Yoshida M,“SIRT2 regulates cell migration through Keap1-mediated cell periphery localization of cortactin”, The RIKEN-Max Planck Joint Research Center for Systems Chemical Biology 3rd Annual Symposium, May 2014, Kreuth, Germany

伊藤昭博、工藤幹雄、吉田稔、「アロステリックなSirtuin阻害剤の発見」、2014年度農芸化学会大会、2014年3月、川崎

伊藤昭博、「タンパク質SUMO化を標的とした化合物の同定とその抗がん活性」、新学術領域 天然物ケミカルバイオロジー地区ミニシンポジウム「化学と生物学の打開策 次世代天然物ケミカルバイオロジー研究の開拓へ」、2014年3月、和光 招待講演

Ito A, Maeda S, Yoshida M,“Regulation of cortactin-mediated cell migration by Keap1 and SIRT2”, The 7th Japan-Korea Chemical Biology Symposium, February 2014, Jeju Island, Korea

Ito A, “Identification of novel histone H3K9 methyltransferase inhibitors using a fluorescent based-screening system and visualization of histone H3K9 methylation in living cells”, The 9th AFMC International Medicinal Chemistry Symposium, October 2013, Taipei, Taiwan 招待講演

伊藤昭博、吉田稔、「Keap1によるcortactinの細胞内局在調節を介した細胞運動の制御機構」、第72回日本癌学会学術総会、2013年10月、横浜

伊藤昭博、吉田稔、「タンパク質SUMO化を標的とした化合物の探索」、第86回日本生化学会大会、2013年9月、横浜 招待講演

Maemoto Y, Kiso S, Shibata H and Maki M. Analyses of ESCRT-dependent limited proteolytic activities of calpain-7 in mammalian cells. FASEB Science Research Conference “The Biology of Calpains in Health and Disease” July 2013, Saxtons River, Vermont

伊藤昭博、掛谷秀昭、吉田稔、「脱SUMO化酵素SENP1阻害剤の探索」、第17回日本がん分子標的治療学会学術集会、2013年6月

Ito A, Maeda S, Yoshida M,“Keap1 and SIRT2 regulate the cell migration by controlling the cortactin activity, The Riken-Max Planck Joint Research Center for systems chemical biology the second symposium, April 2013, Wako, Japan

伊藤昭博、「SUMO化・脱SUMO化阻害剤の現状」、第10回SUMO化研究会、2012年11月、熊本

伊藤昭博、「ヒストンリジンメチル化酵素Set7/9阻害剤の発見」、PerkinElmer 75周年記念イベント For the Better Forum 2012、2012年11月、東京 招待講演

伊藤昭博、木村賢一、吉田稔、「SUMO E2阻害剤としてのエラグ酸の発見とがん治療薬としての可能性」、第71回日本癌学会学術総会、2012年9月、札幌

Ito A, Takahashi S, Yoshida M, “Reversible acetylation regulates ketogenesis in response to fasting by modulating the HMG-CoA lyase activity”, New Frontiers of Metabolism Research in Biomedical Sciences, September 2012, Tokyo, Japan.

Ito A, “Regulation of energy metabolism and cancer cell motility by protein acetylation”, Core-to-Core Project sponsored by JSPS “New Insights into the Molecular Basis of Prevention of Diseases in the Aging Society Caused by Modulation of Insulin-Like Activities”(FY2012-FY2017)Open International Seminar, August 2012, Tokyo, Japan

Ito A, Hirohama M, Igarashi Y, Kondoh Y, Saito T, Osada H, Yoshida M, “Spectomycin B1 suppresses Estrogen Receptor α-Dependent Breast Cancer Cell Growth by inhibiting a SUMO E2 Activity”, The 2nd SNU-RIKEN Joint Symposium on Chemical Biology, May 2012, Chuncheon, South Korea

Ito A, Hirohama M, Igarashi Y, Kondoh Y, Saito T, Osada H, Yoshida M, “Discovery of spectomycin B1 as a SUMO E2 inhibitor and its potential for anti-breast cancer therapy”, Riken-Max Planck Joint Research Center kick off symposium, March 2012, Dortmund, Germany

米山操、広浜美香子、木村賢一、半田宏、伊藤昭博、吉田稔、「エラグ酸によるタンパク質SUMO化阻害」、日本農芸化学会2012年度大会、2012年3月、京都

Ito A,“Regulation of mitochondria by protein acetylation, The 3rd INSERM/JSPS meeting-workshop, August 2011, Grenoble, France 口頭発表Ito A, Hirohama M, Igarashi Y, Kondoh Y, Saito T, Osada H, Yoshida M,“Discovery of Spectomycin B1 as a SUMO E2 Inhibitor”, The 6th Korean-Japan Chemical Biology Symposium, January 2012, Sapporo, Japan

伊藤昭博、「SUMO E2阻害剤の発見」、第2回京都大学GCOE-理化学研究所 共同シンポジウム、2011年11月、京都

伊藤昭博、吉田稔、「Real-time imaging of histone H4K12 acetylationand evaluation of anticancer drugs using a novel fluorescent probe」、第70回日本癌学会学術総会、2011年10月、名古屋

伊藤昭博、「タンパク質SUMO化を標的とした小分子化合物の探索」、第9回SUMO研究会、2011年9月、出雲

伊藤昭博、吉田稔、「Spectomycin B1はSUMO E2を標的とする新規SUMO化阻害剤である」、日本がん分子標的治療学会 第15回学術集会、2011年6月、東京

Ito A, Maeda S, Yoshida M,“Regulation of the cortactin activity by acetylation and Keap1, The 1st RIKEN -SNU workshop on Chemistry and Biology, March 2011, Nagatoro-cho, Japan

Maemoto Y, Osako Y, Tanaka R, Suzuki H, Shibata H and Maki M. ESCRT-related protein IST1 interacts with human calpain-7 and enhances its autolytic activity. FASEB Science Research Conference “The Biology of Calpains in Health and Disease” July 2010, Carefree, Arizona

Books

Ito A and Yoshida M, "Epigenetics: Bioprobes that target epigenetic modifications", Bioprobes: Biochemical Tools for Investigating Cell Function, 37-74, 2017

伊藤昭博、工藤紀雄、吉田稔、「コータクチンのアセチル化によるがん細胞運動制御機構と創薬研究」、MEDCHEM NEWS 27, 190-194, 2017

伊藤昭博、中田明子、吉田稔、「HDAC活性・HAT活性測定法」、実験医学(別冊):エピジェネティクス実験スタンダード もう悩まない! ゲノム機能制御の読み解き方、212-221, 2017

伊藤昭博、吉田稔、「エピジェネティク創薬スクリーニング」、エピゲノム研究 修飾の全体像の理解から先制・個別化医療へ 実験医学増刊 34, 178-184, 2016

伊藤昭博、八代田陽子、吉田稔、「NAD+関連タンパク質」 細胞シグナル操作法 生体の科学 66, 454-455, 2015

小林大貴、伊藤昭博、「エピジェネティクに基づく技術開発と創薬」、アレルギーとエピジェネティクス アレルギー・免疫 医薬ジャーナル社 21, 28-35, 2014

伊藤昭博、小林大貴、吉田稔、「ヒストン修飾酵素を標的としたエピジェネティクス創薬」、エピジェネティクスの産業応用 シーエムシー出版 319-332, 2014

伊藤昭博、小林大貴、吉田稔、「エピジェネティクス修飾を標的とする阻害剤」、エピジェネティクスキーワード事典 羊土社 290-301 (2013)

伊藤昭博、吉田稔、「発がんに関わるヒストン修飾酵素を標的とした抗がん剤の開発」、がん基礎性物学 —革新的シーズ育成に向けてー 220-226, 2013

伊藤昭博、吉田稔、「エネルギー代謝を制御する脱アセチル化酵素サーチュインのケミカルバイオロジー」、内分泌・糖尿病・代謝内科6、508-514, 2012

伊藤昭博、工藤幹雄、吉田稔、「ヒストン脱アセチル化酵素阻害剤とがん治療への応用」、造血器腫瘍とエピジェネティクス —治療への応用と新たな展開—、医薬ジャーナル社、40-48, 2012

伊藤昭博、吉田稔、「ヒストン脱アセチル化酵素」、分子標的薬-がんから他疾患までの治癒をめざして-、日本臨牀社 増刊号、78-83, 2012

竹本靖、伊藤昭博、吉田稔、「エピジェネティクス制御化合物」、分子標的薬-がんから他疾患までの治癒をめざして-、日本臨牀社 増刊号、403-406(2012)

伊藤昭博、「ヒストンH4の化学修飾」、生体の科学 特集 細胞核―構造と機能 62, 444-445, 2011

松田(古園)さおり、伊藤昭博、吉田稔、「アセチローム解析から見えてきたタンパク質修飾の新しい意味-ヒストン修飾を超えて、代謝・細胞機能に及ぼす作用」、実験医学29, 2153-2158 , 2011

伊藤環、伊藤昭博、吉田稔、「ケミカルエピジェネティクス-化合物によるエピジェネティクス情報の制御」、実験医学 28, 122-130, 2010

伊藤昭博、吉田稔、「タンパク質SUMO化阻害剤の発見」、バイオサイエンスとインダストリー 67, 347-349, 2009

竹本靖、伊藤昭博、福田勲、吉田稔、「創薬標的としてのエピジェネティクス制御機構」、実験医学(増刊):分子標的薬開発への新たなる挑戦 27, 204-212, 2009

伊藤昭博、前田里子、吉田稔、「HAT活性・HDAC活性測定」、実験医学別冊 エピジェネティクス実験プロトコール 167-180, 2009

Ito A, Nishino N, Yoshida M, “HDAC inhibitors: discovery, development, and clinical impacts”, Histone Deacetylases: Transcriptional Regulation and Other Cellular Functions 271-297, 2006

古米亮平、伊藤昭博、吉田稔、「エピジェネティックス制御化合物と創薬」、実験医学増刊 ゲノムワイドに展開するエピジェネティックス医科学 220-225, 2006

伊藤昭博、吉田稔、「タンパク質アセチル化の解析」、実験医学別冊 タンパク質の翻訳後修飾解析プロトコール 154-157, 2006