Network


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

Hotspot


Dive into the research topics where Osamu Ujikawa is active.

Publication


Featured researches published by Osamu Ujikawa.


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.


Bioorganic & Medicinal Chemistry | 2012

A new class of non-thiazolidinedione, non-carboxylic-acid-based highly selective peroxisome proliferator-activated receptor (PPAR) γ agonists: design and synthesis of benzylpyrazole acylsulfonamides.

Kentaro Rikimaru; Takeshi Wakabayashi; Hidenori Abe; Hiroshi Imoto; Tsuyoshi Maekawa; Osamu Ujikawa; Katsuhito Murase; Takanori Matsuo; Mitsuharu Matsumoto; Chisako Nomura; Hiroko Tsuge; Naoto Arimura; Kazutoshi Kawakami; Junichi Sakamoto; Miyuki Funami; Clifford D. Mol; Gyorgy Snell; Kenneth A. Bragstad; Bi-Ching Sang; Douglas R. Dougan; Toshimasa Tanaka; Nozomi Katayama; Yoshiaki Horiguchi; Yu Momose

Herein, we describe the design, synthesis, and structure-activity relationships of novel benzylpyrazole acylsulfonamides as non-thiazolidinedione (TZD), non-carboxylic-acid-based peroxisome proliferator-activated receptor (PPAR) γ agonists. Docking model analysis of in-house weak agonist 2 bound to the reported PPARγ ligand binding domain suggested that modification of the carboxylic acid of 2 would help strengthen the interaction of 2 with the TZD pocket and afford non-carboxylic-acid-based agonists. In this study, we used an acylsulfonamide group as the ring-opening analog of TZD as an isosteric replacement of carboxylic acid moiety of 2; further, preliminary modification of the terminal alkyl chain on the sulfonyl group gave the lead compound 3c. Subsequent optimization of the resulting compound gave the potent agonists 25c, 30b, and 30c with high metabolic stability and significant antidiabetic activity. Further, we have described the difference in binding mode of the carboxylic-acid-based agonist 1 and acylsulfonamide 3d.


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.


ACS Medicinal Chemistry Letters | 2017

Targeting the Allosteric Site of Oncoprotein BCR-ABL as an Alternative Strategy for Effective Target Protein Degradation

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

Protein degradation technology based on hybrid small molecules is an emerging drug modality that has significant potential in drug discovery and as a unique method of post-translational protein knockdown in the field of chemical biology. Here, we report the first example of a novel and potent protein degradation inducer that binds to an allosteric site of the oncogenic BCR-ABL protein. BCR-ABL allosteric ligands were incorporated into the SNIPER (Specific and Nongenetic inhibitor of apoptosis protein [IAP]-dependent Protein Erasers) platform, and a series of in vitro biological assays of binding affinity, target protein modulation, signal transduction, and growth inhibition were carried out. One of the designed compounds, 6 (SNIPER(ABL)-062), showed desirable binding affinities against ABL1, cIAP1/2, and XIAP and consequently caused potent BCR-ABL degradation.


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.


Chemical & Pharmaceutical Bulletin | 2018

Different Degradation Mechanisms of Inhibitor of Apoptosis Proteins (IAPs) by the Specific and Nongenetic IAP-dependent Protein Eraser (SNIPER)

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

Targeted protein degradation by small molecules is an emerging modality with significant potential for drug discovery. We previously developed chimeric molecules, termed specific and non-genetic inhibitor of apoptosis protein (IAP)-dependent protein erasers (SNIPERs), which induce the ubiquitylation and proteasomal degradation of target proteins. This degradation is mediated by the IAPs; the target proteins include bromodomain-containing protein 4 (BRD4), an epigenetic regulator protein. The SNIPER that degrades this particular protein, SNIPER(BRD)-1, consists of an IAP antagonist LCL-161 derivative and a bromodomain and extra-terminal (BET) inhibitor, (+)-JQ-1. SNIPER(BRD)-1 also degrades a cellular inhibitor of apoptosis protein 1 (cIAP1) and an X-linked inhibitor of apoptosis protein (XIAP), the mechanisms of which are not well understood. Here, we show that the degradation of cIAP1 and XIAP by SNIPER(BRD)-1 is induced via different mechanisms. Using a chemical biology-based approach, we developed two inactive SNIPERs, SNIPER(BRD)-3 and SNIPER(BRD)-4, incapable of degrading BRD4. SNIPER(BRD)-3 contained an N-methylated LCL-161 derivative as the IAP ligand, which prevented it from binding IAPs, and resulted in the abrogated degradation of cIAP1, XIAP, and BRD4. SNIPER(BRD)-4, however, incorporated the enantiomer (-)-JQ-1 which was incapable of binding BRD4; this SNIPER degraded cIAP1 but lost the ability to degrade XIAP and BRD4. Furthermore, a mixture of the ligands, (+)-JQ-1 and LCL-161, induced the degradation of cIAP1, but not XIAP and BRD4. These results indicate that cIAP1 degradation is triggered by the binding of the IAP antagonist module to induce autoubiquitylation of cIAP1, whereas a ternary complex formation is required for the SNIPER-induced degradation of XIAP and BRD4.


Archive | 2008

Indazole compounds for activating glucokinase

Tsuneo Yasuma; Shigekazu Sasaki; Osamu Ujikawa; Yasufumi Miyamoto; Stephen L. Gwaltney; Sheldon X. Cao; Andy Jennings


Archive | 2005

Arylalkanoic Acid Derivative

Tsuyoshi Maekawa; Osamu Ujikawa; Hidenori Abe; Izumi Nomura


Archive | 2009

Nitrogenated 5-membered heterocyclic compound

Tsuneo Yasuma; Osamu Ujikawa

Collaboration


Dive into the Osamu Ujikawa's collaboration.

Top Co-Authors

Avatar

Hiroshi Nara

Takeda Pharmaceutical Company

View shared research outputs
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

Katsunori Nagai

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Kenichiro Shimokawa

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Tsuneo Yasuma

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Osamu Sano

Takeda Pharmaceutical Company

View shared research outputs
Researchain Logo
Decentralizing Knowledge