2024/04/27 更新

写真a

ナカジマ ワタル
中嶋 亘
Wataru Nakajima
所属
先端医学研究所 遺伝子制御学部門 講師
職名
講師
外部リンク

研究分野

  • ライフサイエンス / 腫瘍生物学

経歴

  • バージニア州立大学   客員研究員

    2012年10月 - 2014年9月

      詳細を見る

    国名:アメリカ合衆国

    researchmap

論文

  • Loss of p53 function promotes DNA damage-induced formation of nuclear actin filaments. 国際誌

    Takeru Torii, Wataru Sugimoto, Katsuhiko Itoh, Natsuki Kinoshita, Masaya Gessho, Toshiyuki Goto, Ikuno Uehara, Wataru Nakajima, Yemima Budirahardja, Daisuke Miyoshi, Takahito Nishikata, Nobuyuki Tanaka, Hiroaki Hirata, Keiko Kawauchi

    Cell death & disease   14 ( 11 )   766 - 766   2023年11月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Tumor suppressor p53 plays a central role in response to DNA damage. DNA-damaging agents modulate nuclear actin dynamics, influencing cell behaviors; however, whether p53 affects the formation of nuclear actin filaments remains unclear. In this study, we found that p53 depletion promoted the formation of nuclear actin filaments in response to DNA-damaging agents, such as doxorubicin (DOXO) and etoposide (VP16). Even though the genetic probes used for the detection of nuclear actin filaments exerted a promotive effect on actin polymerization, the detected formation of nuclear actin filaments was highly dependent on both p53 depletion and DNA damage. Whilst active p53 is known to promote caspase-1 expression, the overexpression of caspase-1 reduced DNA damage-induced formation of nuclear actin filaments in p53-depleted cells. In contrast, co-treatment with DOXO and the pan-caspase inhibitor Q-VD-OPh or the caspase-1 inhibitor Z-YVAD-FMK induced the formation of nuclear actin filament formation even in cells bearing wild-type p53. These results suggest that the p53-caspase-1 axis suppresses DNA damage-induced formation of nuclear actin filaments. In addition, we found that the expression of nLifeact-GFP, the filamentous-actin-binding peptide Lifeact fused with the nuclear localization signal (NLS) and GFP, modulated the structure of nuclear actin filaments to be phalloidin-stainable in p53-depleted cells treated with the DNA-damaging agent, altering the chromatin structure and reducing the transcriptional activity. The level of phosphorylated H2AX (γH2AX), a marker of DNA damage, in these cells also reduced upon nLifeact-GFP expression, whilst details of the functional relationship between the formation of nLifeact-GFP-decorated nuclear actin filaments and DNA repair remained to be elucidated. Considering that the loss of p53 is associated with cancer progression, the results of this study raise a possibility that the artificial reinforcement of nuclear actin filaments by nLifeact-GFP may enhance the cytotoxic effect of DNA-damaging agents in aggressive cancer cells through a reduction in gene transcription.

    DOI: 10.1038/s41419-023-06310-0

    PubMed

    researchmap

  • The iron chelator deferriferrichrysin induces paraptosis via extracellular signal-related kinase activation in cancer cells. 国際誌

    Natsuki Kinoshita, Masaya Gessho, Takeru Torii, Yukako Ashida, Minori Akamatsu, Alvin Kunyao Guo, Sunmin Lee, Tatsuya Katsuno, Wataru Nakajima, Yemima Budirahardja, Daisuke Miyoshi, Takehiko Todokoro, Hiroki Ishida, Takahito Nishikata, Keiko Kawauchi

    Genes to cells : devoted to molecular & cellular mechanisms   28 ( 9 )   653 - 662   2023年9月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Cancer cells generally exhibit increased iron uptake, which contributes to their abnormal growth and metastatic ability. Iron chelators have thus recently attracted attention as potential anticancer agents. Here, we show that deferriferrichrysin (Dfcy), a natural product from Aspergillus oryzae acts as an iron chelator to induce paraptosis (a programmed cell death pathway characterized by ER dilation) in MCF-7 human breast cancer cells and H1299 human lung cancer cells. We first examined the anticancer efficacy of Dfcy in cancer cells and found that Dfcy induced ER dilation and reduced the number of viable cells. Extracellular signal-related kinase (ERK) was activated by Dfcy treatment, and the MEK inhibitor U0126, a small molecule commonly used to inhibit ERK activity, prevented the increase in ER dilation in Dfcy-treated cells. Concomitantly, the decrease in the number of viable cells upon treatment with Dfcy was attenuated by U0126. Taken together, these results demonstrate that the iron chelator Dfcy exhibits anticancer effects via induction of ERK-dependent paraptosis.

    DOI: 10.1111/gtc.13053

    PubMed

    researchmap

  • Epigenetic Priming with Decitabine Augments the Therapeutic Effect of Cisplatin on Triple-Negative Breast Cancer Cells through Induction of Proapoptotic Factor NOXA. 査読 国際誌

    Wataru Nakajima, Kai Miyazaki, Masahiro Sakaguchi, Yumi Asano, Mariko Ishibashi, Tomoko Kurita, Hiroki Yamaguchi, Hiroyuki Takei, Nobuyuki Tanaka

    Cancers   14 ( 1 )   2022年1月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Epigenetic alterations caused by aberrant DNA methylation have a crucial role in cancer development, and the DNA-demethylating agent decitabine, is used to treat hematopoietic malignancy. Triple-negative breast cancers (TNBCs) have shown sensitivity to decitabine; however, the underlying mechanism of its anticancer effect and its effectiveness in treating TNBCs are not fully understood. We analyzed the effects of decitabine on nine TNBC cell lines and examined genes associated with its cytotoxic effects. According to the effect of decitabine, we classified the cell lines into cell death (D)-type, growth inhibition (G)-type, and resistant (R)-type. In D-type cells, decitabine induced the expression of apoptotic regulators and, among them, NOXA was functionally involved in decitabine-induced apoptosis. In G-type cells, induction of the cyclin-dependent kinase inhibitor, p21, and cell cycle arrest were observed. Furthermore, decitabine enhanced the cytotoxic effect of cisplatin mediated by NOXA in D-type and G-type cells. In contrast, the sensitivity to cisplatin was high in R-type cells, and no enhancing effect by decitabine was observed. These results indicate that decitabine enhances the proapoptotic effect of cisplatin on TNBC cell lines that are less sensitive to cisplatin, indicating the potential for combination therapy in TNBC.

    DOI: 10.3390/cancers14010248

    PubMed

    researchmap

  • Krüppel-Like Factor 4 and Its Activator APTO-253 Induce NOXA-Mediated, p53-Independent Apoptosis in Triple-Negative Breast Cancer Cells. 査読 国際誌

    Wataru Nakajima, Kai Miyazaki, Yumi Asano, Satoshi Kubota, Nobuyuki Tanaka

    Genes   12 ( 4 )   2021年4月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Inducing apoptosis is an effective treatment for cancer. Conventional cytotoxic anticancer agents induce apoptosis primarily through activation of tumor suppressor p53 by causing DNA damage and the resulting regulation of B-cell leukemia/lymphoma-2 (BCL-2) family proteins. Therefore, the effects of these agents are limited in cancers where p53 loss-of-function mutations are common, such as triple-negative breast cancer (TNBC). Here, we demonstrate that ultraviolet (UV) light-induced p53-independent transcriptional activation of NOXA, a proapoptotic factor in the BCL-2 family, results in apoptosis induction. This UV light-induced NOXA expression was triggered by extracellular signal-regulated kinase (ERK) activity. Moreover, we identified the specific UV light-inducible DNA element of the NOXA promoter and found that this sequence is responsible for transcription factor Krüppel-like factor 4 (KLF4)-mediated induction. In p53-mutated TNBC cells, inhibition of KLF4 by RNA interference reduced NOXA expression. Furthermore, treatment of TNBC cells with a KLF4-inducing small compound, APTO-253, resulted in the induction of NOXA expression and NOXA-mediated apoptosis. Therefore, our results help to clarify the molecular mechanism of DNA damage-induced apoptosis and provide support for a possible treatment method for p53-mutated cancers.

    DOI: 10.3390/genes12040539

    PubMed

    researchmap

  • Virus-infection in cochlear supporting cells induces audiosensory receptor hair cell death by TRAIL-induced necroptosis. 査読 国際誌

    Yushi Hayashi, Hidenori Suzuki, Wataru Nakajima, Ikuno Uehara, Atsuko Tanimura, Toshiki Himeda, Satoshi Koike, Tatsuya Katsuno, Shin-Ichiro Kitajiri, Naoto Koyanagi, Yasushi Kawaguchi, Koji Onomoto, Hiroki Kato, Mitsutoshi Yoneyama, Takashi Fujita, Nobuyuki Tanaka

    PloS one   16 ( 11 )   e0260443   2021年

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Although sensorineural hearing loss (SHL) is relatively common, its cause has not been identified in most cases. Previous studies have suggested that viral infection is a major cause of SHL, especially sudden SHL, but the system that protects against pathogens in the inner ear, which is isolated by the blood-labyrinthine barrier, remains poorly understood. We recently showed that, as audiosensory receptor cells, cochlear hair cells (HCs) are protected by surrounding accessory supporting cells (SCs) and greater epithelial ridge (GER or Kölliker's organ) cells (GERCs) against viral infections. Here, we found that virus-infected SCs and GERCs induce HC death via production of the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL). Notably, the HCs expressed the TRAIL death receptors (DR) DR4 and DR5, and virus-induced HC death was suppressed by TRAIL-neutralizing antibodies. TRAIL-induced HC death was not caused by apoptosis, and was inhibited by necroptosis inhibitors. Moreover, corticosteroids, the only effective drug for SHL, inhibited the virus-induced transformation of SCs and GERCs into macrophage-like cells and HC death, while macrophage depletion also inhibited virus-induced HC death. These results reveal a novel mechanism underlying virus-induced HC death in the cochlear sensory epithelium and suggest a possible target for preventing virus-induced SHL.

    DOI: 10.1371/journal.pone.0260443

    PubMed

    researchmap

  • Cochlear supporting cells function as macrophage-like cells and protect audiosensory receptor hair cells from pathogens. 査読 国際誌

    Yushi Hayashi, Hidenori Suzuki, Wataru Nakajima, Ikuno Uehara, Atsuko Tanimura, Toshiki Himeda, Satoshi Koike, Tatsuya Katsuno, Shin-Ichiro Kitajiri, Naoto Koyanagi, Yasushi Kawaguchi, Koji Onomoto, Hiroki Kato, Mitsutoshi Yoneyama, Takashi Fujita, Nobuyuki Tanaka

    Scientific reports   10 ( 1 )   6740 - 6740   2020年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    To protect the audiosensory organ from tissue damage from the immune system, the inner ear is separated from the circulating immune system by the blood-labyrinth barrier, which was previously considered an immune-privileged site. Recent studies have shown that macrophages are distributed in the cochlea, especially in the spiral ligament, spiral ganglion, and stria vascularis; however, the direct pathogen defence mechanism used by audiosensory receptor hair cells (HCs) has remained obscure. Here, we show that HCs are protected from pathogens by surrounding accessory supporting cells (SCs) and greater epithelial ridge (GER or Kölliker's organ) cells (GERCs). In isolated murine cochlear sensory epithelium, we established Theiler's murine encephalomyelitis virus, which infected the SCs and GERCs, but very few HCs. The virus-infected SCs produced interferon (IFN)-α/β, and the viruses efficiently infected the HCs in the IFN-α/β receptor-null sensory epithelium. Interestingly, the virus-infected SCs and GERCs expressed macrophage marker proteins and were eliminated from the cell layer by cell detachment. Moreover, lipopolysaccharide induced phagocytosis of the SCs without cell detachment, and the SCs phagocytosed the bacteria. These results reveal that SCs function as macrophage-like cells, protect adjacent HCs from pathogens, and provide a novel anti-infection inner ear immune system.

    DOI: 10.1038/s41598-020-63654-9

    PubMed

    researchmap

  • DMPK is a new candidate mediator of tumor suppressor p53-dependent cell death 査読

    Katsuhiko Itoh, Takahiro Ebata, Hiroaki Hirata, Takeru Torii, Wataru Sugimoto, Keigo Onodera, Wataru Nakajima, Ikuno Uehara, Daisuke Okuzaki, Shota Yamauchi, Yemima Budirahardja, Takahito Nishikata, Nobuyuki Tanaka, Keiko Kawauchi

    Molecules   24 ( 17 )   2019年9月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    © 2019 by the authors Tumor suppressor p53 plays an integral role in DNA-damage induced apoptosis, a biological process that protects against tumor progression. Cell shape dramatically changes when cells undergo apoptosis, which is associated with actomyosin contraction; however, it remains entirely elusive how p53 regulates actomyosin contraction in response to DNA-damaging agents. To identify a novel p53 regulating gene encoding the modulator of myosin, we conducted DNA microarray analysis. We found that, in response to DNA-damaging agent doxorubicin, expression of myotonic dystrophy protein kinase (DMPK), which is known to upregulate actomyosin contraction, was increased in a p53-dependent manner. The promoter region of DMPK gene contained potential p53-binding sequences and its promoter activity was increased by overexpression of the p53 family protein p73, but, unexpectedly, not of p53. Furthermore, we found that doxorubicin treatment induced p73 expression, which was significantly attenuated by downregulation of p53. These data suggest that p53 induces expression of DMPK through upregulating p73 expression. Overexpression of DMPK promotes contraction of the actomyosin cortex, which leads to formation of membrane blebs, loss of cell adhesion, and concomitant caspase activation. Taken together, our results suggest the existence of p53-p73-DMPK axis which mediates DNA-damage induced actomyosin contraction at the cortex and concomitant cell death.

    DOI: 10.3390/molecules24173175

    Scopus

    PubMed

    researchmap

  • p53-independent Noxa induction by cisplatin is regulated by ATF3/ATF4 in head and neck squamous cell carcinoma cells 査読

    Kanika Sharma, Thien-Trang Vu, Wade Cook, Mitra Naseri, Kevin Zhan, Wataru Nakajima, Hisashi Harada

    Molecular Oncology   12 ( 6 )   788 - 798   2018年6月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:John Wiley and Sons Ltd.  

    The platinum-based DNA damaging agent cisplatin is used as a standard therapy for locally advanced head and neck squamous cell carcinoma (HNSCC). However, the mechanisms underpinning the cytotoxic effects of this compound are not entirely elucidated. Cisplatin produces anticancer effects primarily via activation of the DNA damage response, followed by inducing BCL-2 family dependent mitochondrial apoptosis. We have previously demonstrated that cisplatin induces the expression of proapoptotic BCL-2 family protein, Noxa, that can bind to the prosurvival BCL-2 family protein, MCL-1, to inactivate its function and induce cell death. Here, we show that the upregulation of Noxa is critical for cisplatin-induced apoptosis in p53-null HNSCC cells. This induction is regulated at the transcriptional level. With a series of Noxa promoter-luciferase reporter assays, we find that the CRE (cAMP response element) in the promoter is critical for the Noxa induction by cisplatin treatment. Among the CREB/ATF transcription factors, ATF3 and ATF4 are induced by cisplatin, and downregulation of ATF3 or ATF4 reduced cisplatin-induced Noxa. ATF3 and ATF4 bind to and cooperatively activate the Noxa promoter. Furthermore, ERK1 is involved in cisplatin-induced ATF4 and Noxa induction. In conclusion, ATF3 and ATF4 are important regulators that induce Noxa by cisplatin treatment in a p53-independent manner.

    DOI: 10.1002/1878-0261.12172

    Scopus

    PubMed

    researchmap

  • The anti-apoptotic protein MCL1, a novel target of lung cancer therapy 査読

    Wataru Nakajima, Nobuyuki Tanaka

    Journal of Cancer Treatment and Diagnosis   2   54 - 58   2018年

     詳細を見る

    担当区分:筆頭著者   掲載種別:研究論文(学術雑誌)  

    CiNii Research

    researchmap

  • Chaperone-mediated autophagy promotes lung cancer cell survival through selective stabilization of the pro-survival protein, MCL1 査読

    Junya Suzuki, Wataru Nakajima, Hidenori Suzuki, Yumi Asano, Nobuyuki Tanaka

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   482 ( 4 )   1334 - 1340   2017年1月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Autophagy is a. dynamic recycling system using lysosomal proteolysis that produces new proteins and energy for cellular renovation and homeostasis. Although macroautophagy is known to serve as a survival pathway in many cancer cells, the role of chaperone-mediated autophagy (CMA), a selective protein degradation system, in cancer is not fully understood. Here, we demonstrated that lysosomal proteolysis, but not macroautophagy, attenuated apoptosis induced by the tyrosine kinase inhibitor, crizotinib, in the non-small-cell lung cancer (NSCLC) cell line, EBC1. In EBC1 cells, crizotinib induced BIM-dependent apoptosis, which was enhanced by inhibition of lysosomal proteolysis. Moreover, degradation of the pro-survival protein, MCL1, by the ubiquitin-proteasome system was induced by inhibition of lysosomal proteolysis, and by inhibition of the expression of the CMA mediators, HSC70 (heat shock cognate protein 70 kDa) and LAMP2A (lysosome membrane protein type 2A), suggesting the existence of a CMAmediated MCL1 stabilization system in cancer cells. Indeed, the same MCL1 stabilization system was also observed in several NSCLC cell lines; in these cells, their specific molecular-targeted drug or ABT-263 (Navitoclax), the specific inhibitor of BCL-2 and BCL-X-L, but not of MCL1, effectively induced apoptosis in combination with CMA inhibition. Therefore, our results indicate a novel mechanism of MCL1 stabilization in lung cancers by CMA, and a candidate efficient combination chemotherapy method against lung cancers. (C) 2016 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2016.12.037

    Web of Science

    PubMed

    researchmap

  • DNA damaging agent-induced apoptosis is regulated by MCL-1 phosphorylation and degradation mediated by the Noxa/MCL-1/CDK2 complex 査読

    Wataru Nakajima, Kanika Sharma, June Young Lee, Nicolas T. Maxim, Mark A. Hicks, Thien-Trang Vu, Angela Luu, W. Andrew Yeudall, Nobuyuki Tanaka, Hisashi Harada

    ONCOTARGET   7 ( 24 )   36353 - 36365   2016年6月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:IMPACT JOURNALS LLC  

    Noxa, a BH3-only pro-apoptotic BCL-2 family protein, causes apoptosis by specifically interacting with the anti-apoptotic protein MCL-1 to induce its proteasome-mediated degradation. We show here that the DNA damaging agents cisplatin and etoposide upregulate Noxa expression, which is required for the phosphorylation of MCL-1 at Ser64/Thr70 sites, proteasome-dependent degradation, and apoptosis. Noxa-induced MCL-1 phosphorylation at these sites occurs at the mitochondria and is primarily regulated by CDK2. MCL-1 and CDK2 form a stable complex and Noxa binds to this complex to facilitate the phosphorylation of MCL-1. When Ser64 and Thr70 of MCL-1 are substituted with alanine, the mutated MCL-1 is neither phosphorylated nor ubiquitinated, and becomes more stable than the wild-type protein. As a consequence, this mutant can inhibit apoptosis induced by Noxa overexpression or cisplatin treatment. These results indicate that Noxa-mediated MCL-1 phosphorylation followed by MCL-1 degradation is critical for apoptosis induced by DNA damaging agents through regulation of the Noxa/MCL-1/CDK2 complex.

    DOI: 10.18632/oncotarget.9217

    Web of Science

    PubMed

    researchmap

  • Cisplatin-induced apoptosis in non-small-cell lung cancer cells is dependent on Bax- and Bak-induction pathway and synergistically activated by BH3-mimetic ABT-263 in p53 wild-type and mutant cells 査読

    Masaru Matsumoto, Wataru Nakajima, Masahiro Seike, Akihiko Gemma, Nobuyuki Tanaka

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   473 ( 2 )   490 - 496   2016年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Cisplatin is a highly effective anticancer drug for treatment of various tumors including non-small-cell lung cancer (NSCLC), and is especially useful in cases nonresponsive to molecular-targeted drugs. Accumulating evidence has shown that cisplatin activates the p53-dependent apoptotic pathway, but it also induces apoptosis in p53-mutated cancer cells. Here we demonstrated that DNA-damage inducible proapoptotic BH3 (Bcl-2 homology region 3)-only Bcl-2 family members, Noxa, Puma, Bim and Bid, are not involved in cisplatin-induced apoptosis in human NSCLC cell lines. In contrast, the expression of proapoptotic multidomain Bcl-2-family members, Bak and Bax, was induced by cisplatin in p53 dependent and-independent manners, respectively. Moreover, in wild-type p53-expressing cells, cisplatin mainly used the Bak-dependent apoptotic pathway, but this apoptotic pathway shifted to the Bax-dependent pathway by loss-of-function of p53. Furthermore, both Bak- and Bax-induced apoptosis was enhanced by the antiapoptotic Bcl-2 family member, Bcl-X-L knockdown, but not by Mcl-1 knockdown. From this result, we tested the effect of ABT-263 (Navitoclax), the specific inhibitor of Bcl-2 and Bcl-XL, but not Mcl-1, and found that ABT-263 synergistically enhanced cisplatin-induced apoptosis in NSCLC cells in the presence or absence of p53. These results indicate a novel regulatory system in cisplatin-induced NSCLC cell apoptosis, and a candidate efficient combination chemotherapy method against lung cancers. (C) 2016 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2016.03.053

    Web of Science

    PubMed

    researchmap

  • Exploitation of the Apoptosis-Primed State of MYCN-Amplified Neuroblastoma to Develop a Potent and Specific Targeted Therapy Combination 査読

    Jungoh Ham, Carlotta Costa, Renata Sano, Timothy L. Lochmann, Erin M. Sennott, Neha U. Patel, Anahita Dastur, Maria Gomez-Caraballo, Kateryna Krytska, Aaron N. Hata, Konstantinos V. Floros, Mark T. Hughes, Charles T. Jakubik, Daniel A. R. Heisey, Justin T. Ferrell, Molly L. Bristol, Ryan J. March, Craig Yates, Mark A. Hicks, Wataru Nakajima, Madhu Gowda, Brad E. Windle, Mikhail G. Dozmorov, Mathew J. Garnett, Ultan McDermott, Hisashi Harada, Shirley M. Taylor, Iain M. Morgan, Cyril H. Benes, Jeffrey A. Engelman, Yael P. Mosse, Anthony C. Faber

    CANCER CELL   29 ( 2 )   159 - 172   2016年2月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:CELL PRESS  

    Fewer than half of children with high-risk neuroblastoma survive. Many of these tumors harbor high-level amplification of MYCN, which correlates with poor disease outcome. Using data from our large drug screen we predicted, and subsequently demonstrated, that MYCN-amplified neuroblastomas are sensitive to the BCL-2 inhibitor ABT-199. This sensitivity occurs in part through low anti-apoptotic BCL-xL expression, high pro-apoptotic NOXA expression, and paradoxical, MYCN-driven upregulation of NOXA. Screening for enhancers of ABT-199 sensitivity in MYCN-amplified neuroblastomas, we demonstrate that the Aurora Kinase A inhibitor MLN8237 combines with ABT-199 to induce widespread apoptosis. In diverse models of MYCN-amplified neuroblastoma, including a patient-derived xenograft model, this combination uniformly induced tumor shrinkage, and in multiple instances led to complete tumor regression.

    DOI: 10.1016/j.ccell.2016.01.002

    Web of Science

    PubMed

    researchmap

  • Combination with vorinostat overcomes ABT-263 (navitoclax) resistance of small cell lung cancer 査読

    Wataru Nakajima, Kanika Sharma, Mark A. Hicks, Ngoc Le, Rikiara Brown, Geoffrey W. Krystal, Hisashi Harada

    CANCER BIOLOGY & THERAPY   17 ( 1 )   27 - 35   2016年

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:TAYLOR & FRANCIS INC  

    Small cell lung cancer (SCLC) is an aggressive tumor type with high mortality. One promising approach for SCLC treatment would be to utilize agents targeting molecular abnormalities regulating resistance to apoptosis. BH3 mimetic antagonists, such as ABT-737 and its orally available derivative ABT-263 (navitoclax) have been developed to block the function of pro-survival BCL-2 family members. The sensitivity of SCLC to these drugs varies over a broad range in vitro and in clinical trials. We have previously shown that the expression of Noxa, a BH3-only pro-apoptotic BCL-2 family protein, is a critical determinant of sensitivity to ABT-737. Thus, pharmacological up-regulation of Noxa could enhance cell death induced by the BH3 mimetics. We find that the combination of ABT-263 and a HDAC inhibitor, vorinostat, efficiently induces apoptosis in a variety of SCLC cell lines, including ABT-263 resistant cell lines. Cell death induced by combined treatment is Noxa- and/or BIM-dependent in some cell lines but in others appears to be mediated by down-regulation of BCL-X-L and release of BAK from BCL-X-L and MCL-1. These results suggest that combination of HDAC inhibitors and BCL-2 inhibitors could be an alternative and effective regimen for SCLC treatment.

    DOI: 10.1080/15384047.2015.1108485

    Web of Science

    PubMed

    researchmap

  • Yet Another Function of p53-The Switch That Determines Whether Radiation-Induced Autophagy Will Be Cytoprotective or Nonprotective: Implications for Autophagy Inhibition as a Therapeutic Strategy 査読

    Shweta Chakradeo, Khushboo Sharma, Aisha Alhaddad, Duaa Bakhshwin, Ngoc Le, Hisashi Harada, Wataru Nakajima, W. Andrew Yeudall, Suzy V. Torti, Frank M. Torti, David A. Gewirtz

    MOLECULAR PHARMACOLOGY   87 ( 5 )   803 - 814   2015年5月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER SOC PHARMACOLOGY EXPERIMENTAL THERAPEUTICS  

    The influence of autophagy inhibition on radiation sensitivity was studied in human breast, head and neck, and non-small cell lung cancer cell lines, in cell lines that were either wild type or mutant/null in p53, and in cells where p53 was inducible or silenced. Whereas ionizing radiation promoted autophagy in all tumor cell lines studied, pharmacological inhibition of autophagy and/or genetic silencing of autophagy genes failed to influence sensitivity to radiation in p53 mutant Hs578t breast tumor cells, HN6 head and neck tumor cells, and H358 non-small cell lung cancer cells. The requirement for functional p53 in the promotion of cytoprotective autophagy by radiation was confirmed by the observation that radiation-induced autophagy was nonprotective in p53 null H1299 cells but was converted to the cytoprotective form with induction of p53. Conversely, whereas p53 wild-type HN30 head and neck cancer cells did show sensitization to radiation upon autophagy inhibition, HN30 cells in which p53 was knocked down using small hairpin RNA failed to be sensitized by pharmacological autophagy inhibition. Taken together, these findings indicate that radiation-induced autophagy can be either cytoprotective or nonprotective, a functional difference related to the presence or absence of function p53. Alternatively, these findings could be interpreted to suggest that whereas radiation can induce autophagy independent of p53 status, inhibition of autophagy promotes enhanced radiation sensitivity through a mechanism that requires functional p53. These observations are likely to have direct implications with respect to clinical efforts to modulate the response of malignancies to radiation through autophagy inhibition.

    DOI: 10.1124/mol.114.095273

    Web of Science

    PubMed

    researchmap

  • p53-mediated activation of the mitochondrial protease HtrA2/Omi prevents cell invasion 査読

    Shota Yamauchi, Yan Yan Hou, Alvin Kunyao Guo, Hiroaki Hirata, Wataru Nakajima, Ai Kia Yip, Cheng-han Yu, Ichiro Harada, Keng-Hwee Chiam, Yasuhiro Sawada, Nobuyuki Tanaka, Keiko Kawauchi

    JOURNAL OF CELL BIOLOGY   204 ( 7 )   1191 - 1207   2014年3月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:ROCKEFELLER UNIV PRESS  

    Oncogenic Ras induces cell transformation and promotes an invasive phenotype. The tumor suppressor p53 has a suppressive role in Ras-driven invasion. However, its mechanism remains poorly understood. Here we show that p53 induces activation of the mitochondrial protease high-temperature requirement A2 (HtrA2; also known as Omi) and prevents Ras-driven invasion by modulating the actin cytoskeleton. Oncogenic Ras increases accumulation of p53 in the cytoplasm, which promotes the translocation of p38 mitogen-activated protein kinase (MAPK) into mitochondria and induces phosphorylation of HtrA2/Omi. Concurrently, oncogenic Ras also induces mitochondrial fragmentation, irrespective of p53 expression, causing the release of HtrA2/Omi from mitochondria into the cytosol. Phosphorylated HtrA2/Omi therefore cleaves beta-actin and decreases the amount of filamentous actin (F-actin) in the cytosol. This ultimately down-regulates p130 Crk-associated substrate (p130Cas)-mediated lamellipodia formation, countering the invasive phenotype initiated by oncogenic Ras. Our novel findings provide insights into the mechanism by which p53 prevents the malignant progression of transformed cells.

    DOI: 10.1083/jcb.201309107

    Web of Science

    PubMed

    researchmap

  • Noxa determines localization and stability of MCL-1 and consequently ABT-737 sensitivity in small cell lung cancer 査読

    W. Nakajima, M. A. Hicks, N. Tanaka, G. W. Krystal, H. Harada

    CELL DEATH & DISEASE   5   e1052   2014年2月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:NATURE PUBLISHING GROUP  

    The sensitivity to ABT-737, a prototype BH3 mimetic drug, varies in a broad range in small cell lung cancer (SCLC) cells. We have previously shown that the expression of Noxa, a BH3-only pro-apoptotic BCL-2 family protein, is the critical determinant of ABT-737 sensitivity. We show here that Noxa regulates the localization and stability of MCL-1, an anti-apoptotic member, which results in modulating ABT-737 sensitivity. Mutations in Noxa within the BH3 domain, the carboxyl terminus mitochondrial targeting domain, or of ubiquitinated lysines not only change the localization and stability of Noxa itself but also affect the mitochondrial localization and phosphorylation/ubiquitination status of MCL-1 and consequently modulate sensitivity to ABT-737. Results of studies utilizing these mutant proteins indicate that Noxa recruits MCL-1 from the cytosol to the mitochondria. Translocation of MCL-1 initiates its phosphorylation and subsequent ubiquitination, which triggers proteasome-mediated degradation. The precise regulatory mechanisms of Noxa/MCL-1 expression and stability could provide alternative targets to modulate apoptosis induced by BH3 mimetic drugs or other chemotherapeutic reagents.

    DOI: 10.1038/cddis.2014.6

    Web of Science

    PubMed

    researchmap

  • Paclitaxel-induced apoptosis is BAK-dependent, but BAX and BIM-independent in breast tumor. 査読

    Miller AV, Hicks MA, Nakajima W, Richardson AC, Windle JJ, Harada H

    PloS one   8 ( 4 )   e60685   2013年

  • Noxa induces apoptosis in oncogene-expressing cells through catch-and-release mechanism operating between Puma and Mcl-1 査読

    Wataru Nakajima, Nobuyuki Tanaka

    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS   413 ( 4 )   643 - 648   2011年10月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:ACADEMIC PRESS INC ELSEVIER SCIENCE  

    Tumor suppressor p53 induces apoptosis by transcriptional induction of Noxa and Puma, which encode the proapoptotic BH3-only member of the Bcl-2 family proteins. In the p53-mediated tumor surveillance system, p53 induces apoptosis or replicative senescence in oncogene-expressing cells, resulting in elimination of such cells. In this context, we previously found that Noxa and Puma synergistically induce apoptosis. Here, we found the adenovirus oncogene E1A to induce p53-dependently expression of Puma, but not Noxa. The induced Puma associates with antiapoptotic Bcl-2 protein Mcl-1, accompanied by accumulated Mcl-1 protein on mitochondria. Moreover, E1A also reduces expression of the antiapoptotic Bcl-2 protein Bcl-X(L). In contrast, the DNA-damaging agent adriamycin induces Noxa expression in E1A-expressing cells. Interestingly, Mcl-1 knockdown itself induced apoptosis in E1A-expressing MEFs. Furthermore, Noxa displaced Puma's association with Mcl-1, accompanied by Mcl-1 degradation and apoptosis induction by activating mitochondrial apoptotic executers Bax and Bak. These results suggest that p53-induced apoptosis in oncogene-expressing cells is regulated by differential induction and sequential activation of Noxa and Puma. Accumulated Puma by oncogene enhances susceptibility to apoptosis through "catch" in mitochondria by Mcl-1. Subsequently, in response to DNA-damage, Noxa efficiently induces apoptosis by "release" of Puma from Mcl-1. (C) 2011 Elsevier Inc. All rights reserved.

    DOI: 10.1016/j.bbrc.2011.09.036

    Web of Science

    PubMed

    researchmap

  • Interleukin 6 enhances glycolysis through expression of the glycolytic enzymes hexokinase 2 and 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 査読

    Masaru Ando, Lkuno Uehara, Kayo Kogure, Yumi Asano, Wataru Nakajima, Yoshinori Abe, Keiko Kawauchi, Nobuyuki Tanaka

    Journal of Nippon Medical School   77 ( 2 )   97 - 105   2010年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    Enhanced glycolysis is important for oncogenesis and for the survival and proliferation of cancer cells in the tumor microenvironment. Recent studies have also shown that proinflammatory cytokine signaling, such as that mediated by nuclear factor κB and signal transducer and activator of transcription 3 (STAT3), is important for the generation of inflammation-associated tumors. However, the link between inflammation and enhanced glycolysis has not been identified. In the present study, we found that the proinflammatory cytokine interleukin (IL)-6 enhanced glycolysis in mouse embryonic fibroblasts and human cell lines. Moreover, STAT3 activated by IL-6 enhanced the expression of the glycolytic enzymes hexokinase 2 and 6-phosphofructo-2-kinase/fructose-2,6- bisphosphatase-3 (PFKFB3). Ectopic expression of PFKFB3 enhanced glycolysis, suggesting that the IL-6-STAT3 pathway enhances glycolysis through the induction of these enzymes. Our findings may provide a novel mechanism for inflammation-associated oncogenesis.

    DOI: 10.1272/jnms.77.97

    Scopus

    PubMed

    researchmap

  • 5-aza-2'-deoxycytidine restores proapoptotic function of p53 in cancer cells resistant to p53-induced apoptosis 査読

    Shutaro Yagi, Eri Oda-Sato, Ikuno Uehara, Yumi Asano, Wataru Nakajima, Toshiyuki Takeshita, Nobuyuki Tanaka

    CANCER INVESTIGATION   26 ( 7 )   680 - 688   2008年

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:INFORMA HEALTHCARE  

    The expression of p53-target genes encoding the proapoptotic factor Noxa, but not PUMA, was not induced by p53 in HCT116 and SW480 cells, which show resistance to apoptosis in response to p53 overexpression. The lack of p53 inducibility of Noxa was restored by treatment with the DNA methyltransferase inhibitor 5-Aza-2'-deoxycytidine (5-aza-CdR). Furthermore, p53 induced apoptosis in HCT116 and SW480 cells treated with 5-aza-CdR. Moreover, the inhibition of Noxa expression by RNAi in 5-aza-CdR-treated HCT116 cells resulted in the partial inhibition of p53-induced apoptosis. These results suggest that epigenetic cancer therapy is possible for some cancers in combination with forced p53 activation.

    DOI: 10.1080/07357900701840212

    Web of Science

    PubMed

    researchmap

  • Synergistic induction of apoptosis by p53-inducible Bcl-2 family proteins Noxa and Puma 査読

    Wataru Nakajima, Nobuyuki Tanaka

    Journal of Nippon Medical School   74 ( 2 )   148 - 157   2007年4月

     詳細を見る

    担当区分:筆頭著者   記述言語:英語   掲載種別:研究論文(学術雑誌)  

    One critical tumor-suppressive function of p53 is the induction of apoptosis in oncogene-expressing cells. In this context, p53-inducible genes encoding the BH3-only proteins of the Bcl-2 family, Noxa and Puma, were identified. Gene knockout studies revealed that both Noxa and Puma are involved in apoptosis induction in oncogene-expressing cells. BH3-only proteins induce apoptosis, and activate the downstream apoptosis effectors Bax and Bak. In this study, we found that Noxa and Puma synergistically activate Bax and Bak, and induce apoptosis. Although Noxa activates Bak by inactivating Mcl-1 and Bcl-XL, gene knockdown studies revealed that neither Mcl-1 nor Bcl-XL is involved in this synergism. Moreover, Puma, but not Noxa, directly activated Bax in the absence of Bak, and Noxa enhanced Puma-mediated Bax activation in Bak-deficient cells. These results suggest the existence of a novel regulatory pathway for Noxa-mediated apoptosis. Although we detected synergistic induction of apoptosis by Noxa and Bim, a tumor suppressive transcriptional factor FoxO3-inducible protein, no such synergism was observed for other pairs of BH3-only proteins. Bim and Bid, or Bim and Puma. From these results, it can be considered that p53 carefully controls apoptosis by allowing two molecules to share full ability to induce apoptosis.

    DOI: 10.1272/jnms.74.148

    Scopus

    PubMed

    researchmap

▼全件表示

MISC

▼全件表示

講演・口頭発表等

受賞

  • 優秀ポスター賞

    2016年12月   日本分子生物学会   BRCAness乳癌細胞株の微小管阻害薬パクリタキセルに対する抵抗性機構の解析

     詳細を見る

  • 丸山記念研究助成受賞金受賞

    2016年7月   日本医科大学  

    中嶋 亘

     詳細を見る

  • 日本医科大学優秀論文賞

    2008年9月   日本医科大学  

    中嶋 亘

     詳細を見る

共同研究・競争的資金等の研究課題

  • 分子標的薬耐性肺がんにおける薬剤耐性獲得機構の解明と代謝制御を利用した治療法開発

    研究課題/領域番号:22K08293  2022年4月 - 2025年3月

    日本学術振興会  科学研究費助成事業 基盤研究(C)  基盤研究(C)

    中嶋 亘, 中道 真仁

      詳細を見る

    配分額:4160000円 ( 直接経費:3200000円 、 間接経費:960000円 )

    researchmap

  • 乳癌のサブタイプ別に化学療法の治療効果を決定づける因子の解析と治療予測効果の検討

    研究課題/領域番号:16K11161  2016年4月 - 2020年3月

    日本学術振興会  科学研究費助成事業 基盤研究(C)  基盤研究(C)

    中嶋 亘, 田中 信之

      詳細を見る

    配分額:4810000円 ( 直接経費:3700000円 、 間接経費:1110000円 )

    転移性乳癌に対しては薬物療法が主体となり、特に内分泌療法の効果が期待できないタイプの乳癌では化学療法に頼らざるを得ないのが現状である。化学療法薬の一つである微小管阻害薬は細胞分裂に重要な作用を持つ微小管に異常な重合を促進した後に癌細胞の増殖を停止させることで、細胞死(アポトーシス)を誘導する。しかし、微小管阻害薬により細胞増殖を阻害しても、アポトーシスが誘導されにくいタイプであった癌細胞はやがて薬剤抵抗性を獲得してしまい、再び細胞分裂を繰り返し細胞増殖が進行しその後の治療に難渋することが多い。したがい、微小管阻害薬によるアポトーシス誘導を促すために必要な分子機構を理解することによって、微小管阻害薬の治療効果を高めることにつなげることが本研究の目的である。
    本研究では、乳癌細胞株を用いてアポトーシスに関与する遺伝子群の発現をRNA干渉法でスクリーニングしたところ、アポトーシスを引き起こすのに重要な候補因子を同定することができた。この候補因子を発現抑制した乳癌細胞株では微小管阻害薬処理後のアポトーシスの誘導に顕著な抵抗性を示した。また候補因子の発現量が元来少なかった細胞株では微小管阻害薬に対する感受性が低くアポトーシス誘導が抑制されていたのに対し、候補因子を高発現する細胞株では微小管阻害薬に対する感受性が高いことが明らかとなった。これを実証するために、実際の乳癌患者から採取された組織臨床検体の残余分を用いて、同様にこの候補因子の発現量が微小管阻害薬の治療効果を制御しているのかを検討した。さらには、この候補因子の発現量が低い細胞株では、候補因子がエピジェネティックな制御を受けていることが明らかとなり、この候補因子の転写抑制の状態を解除させることで微小管阻害薬の治療効果を高めることができるのではないかと検討中である。

    researchmap

  • 核内癌抑制因子による遺伝子発現制御システムの解析

    研究課題/領域番号:17054039  2005年 - 2009年

    日本学術振興会  科学研究費助成事業 特定領域研究  特定領域研究

    田中 信之, 上原 郁野, 中嶋 亘, 川内 敬子, 飛梅 圭

      詳細を見る

    配分額:86000000円 ( 直接経費:86000000円 )

    癌細胞がグルコース代謝を主なエネルギー供給源として増殖するが、この代謝の変化が癌細胞の増殖に有利に働いていると考えられている。我々は癌抑制因子p53がグルコース代謝を制御しており、p53の機能が欠損すると転写因子NF-κBの活性化を介してグルコース代謝が亢進し、そのことが癌化に重要であることを発見し、p53の新たな癌抑制機構を明らかにした。

    researchmap