Network


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

Hotspot


Dive into the research topics where Akinori Morita is active.

Publication


Featured researches published by Akinori Morita.


FEBS Letters | 2000

Cleavage and phosphorylation of XRCC4 protein induced by X-irradiation

Yoshihisa Matsumoto; Norio Suzuki; Naoki Namba; Noriko Umeda; Xue-Jun Ma; Akinori Morita; Masanori Tomita; Atsushi Enomoto; Shinobu Serizawa; Kazuya Hirano; Kazuo Sakai; Hideyo Yasuda; Yoshio Hosoi

We report the p35 and p60 forms of XRCC4 protein, appearing in human leukemia MOLT‐4 or U937 cells following X‐irradiation or hyperthermia. p35 appeared in conjunction with the cleavage of DNA‐dependent protein kinase catalytic subunit (DNA‐PKcs) and the fragmentation of internucleosomal DNA, and was suppressed by Ac‐DEVD‐CHO. p35 was also produced in vitro by treating MOLT‐4 cell lysate with recombinant caspases, suggesting that p35 was a caspase‐cleaved fragment of XRCC4 in apoptotic cell death. p60 was sensitive to treatment with phosphatase or wortmannin and was undetectable in M059J cells deficient in DNA‐PKcs. However, p60 was found in ataxia‐telangiectasia cells after irradiation. These results indicated p60 as a phosphorylated form of XRCC4, requiring DNA‐PKcs but not ataxia‐telangiectasia mutated (ATM).


Cancer Letters | 2000

Involvement of SAPK/JNK pathway in X-ray-induced rapid cell death of human T-cell leukemia cell line MOLT-4

Atsushi Enomoto; Norio Suzuki; Kazuya Hirano; Yoshihisa Matsumoto; Akinori Morita; Kazuo Sakai; Hideki Koyama

We found that SAPK/JNK was phosphorylated during X-ray-induced rapid cell death of MOLT-4 cells and that acid Sphingomyelinase inhibitor D609 suppressed the rapid cell death as well as phosphorylation of SAPK/JNK. Also C2-ceramide caused phosphorylation of SAPK/JNK, followed by rapid cell death. Further we isolated X-ray-resistant radiation-hybrid clones from MOLT-4 and 50 Gy irradiated mouse FM3A cells by repeated selections with 3 Gy irradiation. One of them named Rh-1a was found resistant to X-ray- as well as C2-ceramide-induced rapid cell death. Rh-1a cells had mouse DNA but no increase in either mouse or human Bcl-2 determined by Western blotting. Accumulation of p53 after X-irradiation was similarly observed in both parental MOLT-4 and Rh-1a cells. However, contrasting to prolonged and prominent phosphorylated status of SAPK/JNK in MOLT-4 cells, Rh-1a cells exhibited short transient increase and FM3A cells showed no increase of phosphorylated status SAPK/JNK after X-irradiation. Therefore, SAPK/JNK activation is considered important in X-ray-induced rapid cell death or apoptosis of MOLT-4 cells.


ChemBioChem | 2008

BODIPY-based fluorescent redox potential sensors that utilize reversible redox properties of flavin.

Yasuyuki Yamada; Yumiko Tomiyama; Akinori Morita; Masahiko Ikekita; Shin Aoki

Living cells are continuously exposed to various stresses, such as reactive oxygen species (ROS), reactive nitrogen intermediates (RNI), UV light, ionizing radiation, and metal ions. In controlling intracellular redox systems (e.g. , redox potential, E’, of cytosols are 0.28 to 0.22 V vs. SHE at pH 7.0), thiol–disulfide equilibria of intracellular thiols, such as glutathione (GSH) and thioredoxin, play critical roles in controlling the whole redox status and regulating structures and functions of proteins. For example, more than 95% of GSH exists as a reduced form in cells and the depletion of GSH levels induces oxidative stresses. In order to understand the mechanisms involved in oxidative signals, antioxidant defense systems, and related intracellular phenomena, fluorescence sensing systems that respond to environmental redox potential are considered to afford a potentially powerful methodology. However, most fluorescent probes of ROS and RNI irreversibly react with these species and examples of sensors that reversibly respond to thiol–disulfide equilibria have been limited. Our strategy for fluorescence sensing of thiol–disulfide equilibria is to utilize flavins, which are well studied cofactors or photoreceptors utilized in natural flavoproteins, including dehydrogenases, oxygenases, DNA photolyases, and thioredoxin reductases. Recently, we reported an artificial DNA photolyase that consist of a Zn-1,4,7,10tetraazacyclododecane complex as binding site for cis–syn


Cancer Research | 2010

Sodium Orthovanadate Inhibits p53-Mediated Apoptosis

Akinori Morita; Shinichi Yamamoto; Bing Wang; Kaoru Tanaka; Norio Suzuki; Shin Aoki; Azusa Ito; Tomohisa Nanao; Soichiro Ohya; Minako Yoshino; Jin Zhu; Atsushi Enomoto; Yoshihisa Matsumoto; Osamu Funatsu; Yoshio Hosoi; Masahiko Ikekita

Sodium orthovanadate (vanadate) inhibits the DNA-binding activity of p53, but its precise effects on p53 function have not been examined. Here, we show that vanadate exerts a potent antiapoptotic activity through both transcription-dependent and transcription-independent mechanisms relative to other p53 inhibitors, including pifithrin (PFT) alpha. We compared the effects of vanadate to PFTalpha and PFTmicro, an inhibitor of transcription-independent apoptosis by p53. Vanadate suppressed p53-associated apoptotic events at the mitochondria, including the loss of mitochondrial membrane potential, the conformational change of Bax and Bak, the mitochondrial translocation of p53, and the interaction of p53 with Bcl-2. Similarly, vanadate suppressed the apoptosis-inducing activity of a mitochondrially targeted temperature-sensitive p53 in stable transfectants of SaOS-2 cells. In radioprotection assays, which rely on p53, vanadate completely protected mice from a sublethal dose of 8 Gy and partially from a lethal dose of 12 Gy. Together, our findings indicated that vanadate effectively suppresses p53-mediated apoptosis by both transcription-dependent and transcription-independent pathways, and suggested that both pathways must be inhibited to completely block p53-mediated apoptosis.


Inorganic Chemistry | 2010

Design and Synthesis of a Fluorescent Probe for Zn2+, 5,7-Bis(N,N-dimethylaminosulfonyl)-8-hydroxyquinoline-Pendant 1,4,7,10-Tetraazacyclododecane and Zn2+-Dependent Hydrolytic and Zn2+-Independent Photochemical Reactivation of Its Benzenesulfonyl-Caged Derivative

Ryosuke Ohshima; Masanori Kitamura; Akinori Morita; Motoo Shiro; Yasuyuki Yamada; Masahiko Ikekita; Eiichi Kimura; Shin Aoki

We previously reported on a 8-quinolinol-pendant cyclen (L(5)) as a Zn(2+) fluorophore (cyclen = 1,4,7,10-tetraazacyclododecane) and its caged derivative, 8-(benzenesulfonyloxy)-5-(N,N-dimethylaminosulfonyl)quinolin-2-ylmethyl-pendant cyclen (BS-caged-L(5)), which can be reactivated by hydrolysis of benzenesulfonyl group upon complexation with Zn(2+) at neutral pH to give a 1:1 Zn(2+)-L(5) complex (Zn(H(-1)L(5))). We report herein on the synthesis of 5,7-bis(N,N-dimethylaminosulfonyl)-8-hydroxyquinolin-2-ylmethyl-pendant cyclen (L(6)) and its caged derivative (BS-caged-L(6)) for more sensitive and more efficient cell-membrane permeability than those of L(5) and BS-caged-L(5). By potentiometric pH, (1)H NMR, and UV-vis spectroscopic titrations, the deprotonation constants pK(a1)-pK(a6) of H(5)L(6) were determined to be <2, <2, <2, 2.5 +/- 0.1 (for the 8-OH group of the quinoline moiety), 9.7 +/- 0.1, and 10.8 +/- 0.1 at 25 degrees C with I = 0.1 (NaNO(3)). The results of (1)H NMR, potentiometric pH, UV-vis, and fluorescent titrations showed that L(6) rapidly forms a 1:1 complex with Zn(2+) (Zn(H(-1)L(6))), the dissociation constant of which is 50 fM at pH 7.4. The fluorescent emission of Zn(H(-1)L(6)) at 478 nm is 32 times as large as that of L(6) (excitation at 370 nm), and the fluorescent quantum yield of Zn(H(-1)L(6)) (Phi(F) = 0.41) is much greater than that of Zn(H(-1)L(5)) (Phi(F) = 0.044). The BS-caged-L(6) was reactivated by hydrolysis of the benzenesulfonyl moiety more rapidly (completes in 30 min at pH 7.4 at 37 degrees C) than BS-caged-L(5), presumably enabling the practical detection of Zn(2+) in sample solutions and living cells. The photochemical deprotection of BS-caged-L(6) and the cell membrane permeability of L(6) and BS-caged-L(6) are also described.


Cell Death & Differentiation | 2006

Sodium orthovanadate suppresses DNA damage- induced caspase activation and apoptosis by inactivating p53

Akinori Morita; Jin Zhu; Norio Suzuki; Atsushi Enomoto; Yoshihisa Matsumoto; Masanori Tomita; Takahiko Suzuki; Kuni Ohtomo; Yoshio Hosoi

We previously reported that p42/SETβ is a substrate for caspase-7 in irradiated MOLT-4 cells, and that treating the cells with sodium orthovanadate (vanadate) inhibits p42/SETβs caspase-mediated cleavage. Here, we initially found that the inhibitory effect of vanadate was due to the suppression of caspase activation but not of caspase activity. Further investigations revealed that vanadate suppressed upstream of apoptotic events, such as the loss of mitochondrial membrane potential, the conformational change of Bax, and p53 transactivation, although the accumulation, total phosphorylation, and phosphorylation of six individual sites of p53 were not affected. Importantly, vanadate suppressed p53-dependent apoptosis, but not p53-independent apoptosis. Finally, gel-shift and chromatin immunoprecipitation assays conclusively demonstrated that vanadate inhibits the DNA-binding activity of p53. Vanadate is conventionally used as an inhibitor of protein tyrosine phosphatases (PTPs); however, we recommend that the influence of vanadate not only on PTPs but also on p53 be considered before using it.


Biochemical and Biophysical Research Communications | 2003

Caspase-mediated cleavage of JNK during stress-induced apoptosis

Atsushi Enomoto; Norio Suzuki; Akinori Morita; Michihiko Ito; Chang Qing Liu; Yoshihisa Matsumoto; Katsuji Yoshioka; Tadayoshi Shiba; Yoshio Hosoi

The c-Jun N-terminal kinases (JNKs) are a subfamily of the mitogen-activated protein kinases (MAPKs). The JNKs are encoded by three separate genes (jnk1, jnk2, and jnk3), which are spliced alternatively to create 10 JNK isoforms that are either p46 or p54 in size. In this study, we found that the p52 form of JNK emerged in human leukemia MOLT-4 or U937 cells following X-irradiation or heat treatment. The accumulation of p52 coincided with the reduction of p54 JNK. On the other hand, the amounts of p46 JNK did not change by X-irradiation. Induction of the p52 form of JNK also paralleled the appearance of the active form of caspase-3 and was suppressed by a caspase-specific inhibitor, Ac-DEVD-CHO, but not by Ac-YVAD-CHO. In vitro cleavage assays indicated that recombinant human JNK1beta2 and JNK2beta2 were cleaved by caspase-3, and that the mutation of aspartic acid at position 413 of JNK1beta2 or 410 of JNK2beta2 to alanine abolished the cleavage. Altogether, our results demonstrated that p54 JNKs, at least JNK1beta2 and JNK2beta2, were new selective targets of caspases in JNK splicing variants, and suggested that the p52 form could serve as a marker of apoptosis.


International Journal of Radiation Biology | 2001

Involvement of c-Jun NH2-terminal kinase-1 in heat-induced apoptotic cell death of human monoblastic leukaemia U937 cells.

Atsushi Enomoto; Norio Suzuki; C. Liu; Yun Kang; Jin Zhu; S. Serizawa; Yoshihisa Matsumoto; Akinori Morita; Michihiko Ito; Yoshio Hosoi

Purpose: To determine the involvement of c-Jun NH 2 -terminal kinase-1 (JNK1) and possibly of HSP27 in heat-induced apoptosis of human monoblastic leukaemia U937 cells. Materials and methods: Dominant negative JNK1 (APF), in which the phosphorylation sites Thr-Pro-Tyr were changed to Ala-Pro-Phe, was overexpressed in U937 cells. Cell viability and DNA fragmentation were analysed by the erythrosin-B dye exclusion test and by agarose gel electrophoresis, respectively. Expression of activated caspase-9, phosphorylated JNK1, JNK2, p38 and HSP27 was examined by Western blotting. JNK1 kinase assay was also performed using c-Jun as a substrate. Results: Loss of viability, activated cleavage form of caspase-9 and DNA fragmentation were rapid in U937 cells after 44°;C hyperthermia, while overexpression of dominant negative JNK1 interfered with phosphorylation or activation of JNK1 without affecting that of JNK2 or p38/SAPK, and apparently delayed or reduced cleavage and activation of caspase-9, DNA fragmentation and cell death. Heat-induced phosphorylation of HSP27, observed in parental U937 cells, was suppressed and only slightly detectable in jnk1 mutant cells. Conclusions: Prolonged phosphorylation or activation of JNK1 was considered important for heat-induced apoptosis and JNK1 may control the process possibly through phosphorylation of HSP27 and caspase-9 activation in U937 cells.


Oncogene | 2008

Negative regulation of MEKK1/2 signaling by serine-threonine kinase 38 (STK38).

Atsushi Enomoto; N Kido; Michihiko Ito; Akinori Morita; Yoshihisa Matsumoto; Nobuhiko Takamatsu; Yoshio Hosoi; Kiyoshi Miyagawa

Mitogen-activated protein kinases (MAPKs) are activated through the kinase cascades of MAPK, MAPK kinase (MAPKK) and MAPKK kinase (MAPKKK). MAPKKKs phosphorylate and activate their downstream MAPKKs, which in turn phosphorylate and activate their downstream MAPKs. MAPKKK proteins relay upstream signals through the MAPK cascades to induce cellular responses. However, the molecular mechanisms by which given MAPKKKs are regulated remain largely unknown. Here, we found that serine-threonine protein kinase 38, STK38, physically interacts with the MAPKKKs MEKK1 and MEKK2 (MEKK1/2). The carboxy terminus, including the catalytic domain, but not the amino terminus of MEKK1/2 was necessary for the interaction with STK38. STK38 inhibited MEKK1/2 activation without preventing MEKK1/2 binding to its substrate, SEK1. Importantly, STK38 suppressed the autophosphorylation of MEKK2 without interfering with MEKK2 dimer formation, and converted MEKK2 from its phosphorylated to its nonphosphorylated form. The negative regulation of MEKK1/2 was not due to its phosphorylation by STK38. On the other hand, stk38 short hairpin RNA enhanced sorbitol-induced activation of MEKK2 and phosphorylation of the downstream MAPKKs, MKK3/6. Taken together, our results indicate that STK38 negatively regulates the activation of MEKK1/2 by direct interaction with the catalytic domain of MEKK1/2, suggesting a novel mechanism of MEKK1/2 regulation.


Bioorganic & Medicinal Chemistry Letters | 2011

3-(3-Phenoxybenzyl)amino-β-carboline: A novel antitumor drug targeting α-tubulin

Reiko Ikeda; Masaki Kurosawa; Takazumi Okabayashi; Ayako Takei; Masamichi Yoshiwara; Tadashi Kumakura; Norio Sakai; Osamu Funatsu; Akinori Morita; Masahiko Ikekita; Yumi Nakaike; Takeo Konakahara

3-(3-Phenoxybenzyl)amino-β-carboline 2h showed extremely-high activity; the IC(50) value was 0.074 μM. To verify 2h-induced cell death types, we observed the chromatin condensation, the DNA fragmentation and activated caspase-3 using Hoechst 33342, agarose electrophoresis and western blot, and suggesting 2h-induced cell death type was apoptosis. Flow cytometry showed that 2h-treated cell was induced SubG1 cell population after G2/M cell cycle arrest. In addition, using affinity chromatography and peptide mass fingerprinting, we found that interacting protein with this compound was α-tubulin protein.

Collaboration


Dive into the Akinori Morita's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yoshihisa Matsumoto

Tokyo Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bing Wang

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar

Masahiko Ikekita

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar

Shin Aoki

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar

Kaoru Tanaka

National Institute of Radiological Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge