Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Katsunori Nagai is active.

Publication


Featured researches published by Katsunori Nagai.


Cell Death and Disease | 2014

Cancer cell death induced by novel small molecules degrading the TACC3 protein via the ubiquitin–proteasome pathway

Nobumichi Ohoka; Katsunori Nagai; Takayuki Hattori; Keiichiro Okuhira; Norihito Shibata; Nobuo Cho; Mikihiko Naito

The selective degradation of target proteins with small molecules is a novel approach to the treatment of various diseases, including cancer. We have developed a protein knockdown system with a series of hybrid small compounds that induce the selective degradation of target proteins via the ubiquitin–proteasome pathway. In this study, we designed and synthesized novel small molecules called SNIPER(TACC3)s, which target the spindle regulatory protein transforming acidic coiled-coil-3 (TACC3). SNIPER(TACC3)s induce poly-ubiquitylation and proteasomal degradation of TACC3 and reduce the TACC3 protein level in cells. Mechanistic analysis indicated that the ubiquitin ligase APC/CCDH1 mediates the SNIPER(TACC3)-induced degradation of TACC3. Intriguingly, SNIPER(TACC3) selectively induced cell death in cancer cells expressing a larger amount of TACC3 protein than normal cells. These results suggest that protein knockdown of TACC3 by SNIPER(TACC3) is a potential strategy for treating cancers overexpressing the TACC3 protein.


Tetrahedron Letters | 1997

SOLID PHASE SYNTHESIS OF 2-AMINOBUTADIENES USING A PIPERAZINE LINKER

Nicholas W. Hird; Kazuyuki Irie; Katsunori Nagai

Abstract A series of resin-bound 4-substituted-2-aminobutadienes have been synthesised via Wittig reaction with polymer supported 2-( N -piperazino)prop-1-enyl-1-triphenylphosphonium bromide. The use of piperazine provides a readily cleavable enamine linker for attachment of ketones that is compatible with anion chemistry.


Journal of Biological Chemistry | 2017

In Vivo Knockdown of Pathogenic Proteins via Specific and Nongenetic IAP-dependent Protein Erasers (SNIPERs)

Nobumichi Ohoka; Keiichiro Okuhira; Masahiro Ito; Katsunori Nagai; Norihito Shibata; Takayuki Hattori; Osamu Ujikawa; Kenichiro Shimokawa; Osamu Sano; Ryokichi Koyama; Hisashi Fujita; Mika Teratani; Hirokazu Matsumoto; Yasuhiro Imaeda; Hiroshi Nara; Nobuo Cho; Mikihiko Naito

Many diseases, especially cancers, result from aberrant or overexpression of pathogenic proteins. Specific inhibitors against these proteins have shown remarkable therapeutic effects, but these are limited mainly to enzymes. An alternative approach that may have utility in drug development relies on selective degradation of pathogenic proteins via small chimeric molecules linking an E3 ubiquitin ligase to the targeted protein for proteasomal degradation. To this end, we recently developed a protein knockdown system based on hybrid small molecule SNIPERs (Specific and Nongenetic IAP-dependent Protein Erasers) that recruit inhibitor of the apoptosis protein (IAP) ubiquitin ligases to specifically degrade targeted proteins. Here, we extend our previous study to show a proof of concept of the SNIPER technology in vivo. By incorporating a high affinity IAP ligand, we developed a novel SNIPER against estrogen receptor α (ERα), SNIPER(ER)-87, that has a potent protein knockdown activity. The SNIPER(ER) reduced ERα levels in tumor xenografts and suppressed the growth of ERα-positive breast tumors in mice. Mechanistically, it preferentially recruits X-linked IAP (XIAP) rather than cellular IAP1, to degrade ERα via the ubiquitin-proteasome pathway. With this IAP ligand, potent SNIPERs against other pathogenic proteins, BCR-ABL, bromodomain-containing protein 4 (BRD4), and phosphodiesterase-4 (PDE4) could also be developed. These results indicate that forced ubiquitylation by SNIPERs is a useful method to achieve efficient protein knockdown with potential therapeutic activities and could also be applied to study the role of ubiquitylation in many cellular processes.


Tetrahedron Letters | 1997

Synthesis of 3,4,5-trisubstituted cyclohexanones by cycloaddition to solid phase 2-aminobutadienes

Melanie Crawshaw; Nicholas W. Hird; Kazuyuki Irie; Katsunori Nagai

Abstract [4+2] Cycloaddition of maleimides and nitrostyrenes to resin bound 4-substituted-2-aminobutadienes gives, after cleavage, 3,4,5-trisubstituted cyclohexanones in moderate to good yields and high purities. The reaction occurs under mild conditions and has been automated on the ACT 496 synthesiser.


Cancer Science | 2017

Development of protein degradation inducers of oncogenic BCR‐ABL protein by conjugation of ABL kinase inhibitors and IAP ligands

Norihito Shibata; Naoki Miyamoto; Katsunori Nagai; Kenichiro Shimokawa; Tomoya Sameshima; Nobumichi Ohoka; Takayuki Hattori; Yasuhiro Imaeda; Hiroshi Nara; Nobuo Cho; Mikihiko Naito

Chromosomal translocation occurs in some cancer cells, which results in the expression of aberrant oncogenic fusion proteins that include BCR‐ABL in chronic myelogenous leukemia (CML). Inhibitors of ABL tyrosine kinase, such as imatinib and dasatinib, exhibit remarkable therapeutic effects, although emergence of drug resistance hampers the therapy during long‐term treatment. An alternative approach to treat CML is to downregulate the BCR‐ABL protein. We have devised a protein knockdown system by hybrid molecules named Specific and Non‐genetic inhibitor of apoptosis protein [IAP]‐dependent Protein Erasers (SNIPER), which is designed to induce IAP‐mediated ubiquitylation and proteasomal degradation of target proteins, and a couple of SNIPER(ABL) against BCR‐ABL protein have been developed recently. In this study, we tested various combinations of ABL inhibitors and IAP ligands, and the linker was optimized for protein knockdown activity of SNIPER(ABL). The resulting SNIPER(ABL)‐39, in which dasatinib is conjugated to an IAP ligand LCL161 derivative by polyethylene glycol (PEG) × 3 linker, shows a potent activity to degrade the BCR‐ABL protein. Mechanistic analysis suggested that both cellular inhibitor of apoptosis protein 1 (cIAP1) and X‐linked inhibitor of apoptosis protein (XIAP) play a role in the degradation of BCR‐ABL protein. Consistent with the degradation of BCR‐ABL protein, the SNIPER(ABL)‐39 inhibited the phosphorylation of signal transducer and activator of transcription 5 (STAT5) and Crk like proto‐oncogene (CrkL), and suppressed the growth of BCR‐ABL‐positive CML cells. These results suggest that SNIPER(ABL)‐39 could be a candidate for a degradation‐based novel anti‐cancer drug against BCR‐ABL‐positive CML.


Journal of Medicinal Chemistry | 2017

Development of Protein Degradation Inducers of Androgen Receptor by Conjugation of Androgen Receptor Ligands and Inhibitor of Apoptosis Protein Ligands

Norihito Shibata; Katsunori Nagai; Yoko Morita; Osamu Ujikawa; Nobumichi Ohoka; Takayuki Hattori; Ryokichi Koyama; Osamu Sano; Yasuhiro Imaeda; Hiroshi Nara; Nobuo Cho; Mikihiko Naito

Targeted protein degradation using small molecules is a novel strategy for drug development. We have developed hybrid molecules named specific and nongenetic inhibitor of apoptosis protein [IAP]-dependent protein erasers (SNIPERs) that recruit IAP ubiquitin ligases to degrade target proteins. Here, we show novel SNIPERs capable of inducing proteasomal degradation of the androgen receptor (AR). Through derivatization of the SNIPER(AR) molecule at the AR ligand and IAP ligand and linker, we developed 42a (SNIPER(AR)-51), which shows effective protein knockdown activity against AR. Consistent with the degradation of the AR protein, 42a inhibits AR-mediated gene expression and proliferation of androgen-dependent prostate cancer cells. In addition, 42a efficiently induces caspase activation and apoptosis in prostate cancer cells, which was not observed in the cells treated with AR antagonists. These results suggest that SNIPER(AR)s could be leads for an anticancer drug against prostate cancers that exhibit AR-dependent proliferation.


Cancer Science | 2017

SNIPER(TACC3) induces cytoplasmic vacuolization and sensitizes cancer cells to Bortezomib

Nobumichi Ohoka; Katsunori Nagai; Norihito Shibata; Takayuki Hattori; Hiroshi Nara; Nobuo Cho; Mikihiko Naito

We previously developed a hybrid small molecule SNIPER (Specific and Nongenetic IAP‐dependent Protein ERaser) against transforming acidic coiled‐coil‐3 (TACC3), SNIPER(TACC3), that induces proteasomal degradation of TACC3 protein. In this study, we found that SNIPER(TACC3) induces cytoplasmic vacuolization derived from endoplasmic reticulum (ER) and paraptosis‐like cell death selectively in cancer cells. Mechanistic analysis suggests that accumulation of ubiquitylated protein aggregates that requires X‐linked inhibitor of apoptosis protein (XIAP) induces ER stress, which results in ER‐stress responses involving X‐box binding protein‐1 (XBP‐1) and ER‐derived vacuolization in cancer cells. Importantly, inhibition of proteasome enhanced the SNIPER(TACC3)‐induced vacuolization, and the combination treatment of SNIPER(TACC3) and bortezomib exhibited a synergistic anticancer activity in several cancer cell lines. The induction of paraptosis‐like cell death in cancer cells by SNIPER(TACC3) could be applied to treat cancer cells resistant to undergo apoptosis by overexpression of XIAP.


Journal of Biological Chemistry | 2018

Derivatization of inhibitor of apoptosis protein (IAP) ligands yields improved inducers of estrogen receptor α degradation

Nobumichi Ohoka; Yoko Morita; Katsunori Nagai; Kenichiro Shimokawa; Osamu Ujikawa; Ikuo Fujimori; Masahiro Ito; Youji Hayase; Keiichiro Okuhira; Norihito Shibata; Takayuki Hattori; Tomoya Sameshima; Osamu Sano; Ryokichi Koyama; Yasuhiro Imaeda; Hiroshi Nara; Nobuo Cho; Mikihiko Naito

Aberrant expression of proteins often underlies many diseases, including cancer. A recently developed approach in drug development is small molecule-mediated, selective degradation of dysregulated proteins. We have devised a protein-knockdown system that utilizes chimeric molecules termed specific and nongenetic IAP-dependent protein erasers (SNIPERs) to induce ubiquitylation and proteasomal degradation of various target proteins. SNIPER(ER)-87 consists of an inhibitor of apoptosis protein (IAP) ligand LCL161 derivative that is conjugated to the estrogen receptor α (ERα) ligand 4-hydroxytamoxifen by a PEG linker, and we have previously reported that this SNIPER efficiently degrades the ERα protein. Here, we report that derivatization of the IAP ligand module yields SNIPER(ER)s with superior protein-knockdown activity. These improved SNIPER(ER)s exhibited higher binding affinities to IAPs and induced more potent degradation of ERα than does SNIPER(ER)-87. Further, they induced simultaneous degradation of cellular inhibitor of apoptosis protein 1 (cIAP1) and delayed degradation of X-linked IAP (XIAP). Notably, these reengineered SNIPER(ER)s efficiently induced apoptosis in MCF-7 human breast cancer cells that require IAPs for continued cellular survival. We found that one of these molecules, SNIPER(ER)-110, inhibits the growth of MCF-7 tumor xenografts in mice more potently than the previously characterized SNIPER(ER)-87. Mechanistic analysis revealed that our novel SNIPER(ER)s preferentially recruit XIAP, rather than cIAP1, to degrade ERα. Our results suggest that derivatized IAP ligands could facilitate further development of SNIPERs with potent protein-knockdown and cytocidal activities against cancer cells requiring IAPs for survival.


Scientific Reports | 2018

Pharmacological difference between degrader and inhibitor against oncogenic BCR-ABL kinase

Norihito Shibata; Kenichiro Shimokawa; Katsunori Nagai; Nobumichi Ohoka; Takayuki Hattori; Naoki Miyamoto; Osamu Ujikawa; Tomoya Sameshima; Hiroshi Nara; Nobuo Cho; Mikihiko Naito

Chronic myelogenous leukemia (CML) is characterized by the oncogenic fusion protein, BCR-ABL protein kinase, against which clinically useful inhibitors have been developed. An alternative approach to treat CML is to degrade the BCR-ABL protein. Recently, potent degraders against BCR-ABL have been developed by conjugating dasatinib to ligands for E3 ubiquitin ligases. Since the degraders contain the dasatinib moiety, they also inhibit BCR-ABL kinase activity, which complicates our understanding of the impact of BCR-ABL degradation by degraders in CML growth inhibition. To address this issue, we chose DAS-IAP, as a potent BCR-ABL degrader, and developed a structurally related inactive degrader, DAS-meIAP, which inhibits kinase activity but does not degrade the BCR-ABL protein. DAS-IAP showed slightly weaker activity than DAS-meIAP in inhibiting cell growth when CML cells were treated for 48 h. However, DAS-IAP showed sustained growth inhibition even when the drug was removed after short-term treatment, whereas CML cell growth rapidly resumed following removal of DAS-meIAP and dasatinib. Consistently, suppression of BCR-ABL levels and downstream kinase signaling were maintained after DAS-IAP removal, whereas kinase signaling rapidly recovered following removal of DAS-meIAP and dasatinib. These results indicate that BCR-ABL degrader shows more sustained inhibition of CML cell growth than ABL kinase inhibitor.


Archive | 1997

Carbapenem compound, its production and medicine

Takashi Ichikawa; Tetsuo Miwa; Katsunori Nagai; 隆史 一川; 哲生 三輪; 克典 永井

Collaboration


Dive into the Katsunori Nagai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nobuo Cho

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiroshi Nara

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Osamu Ujikawa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Yasuhiro Imaeda

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kenichiro Shimokawa

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge