Network


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

Hotspot


Dive into the research topics where Yukitoshi Nagahara is active.

Publication


Featured researches published by Yukitoshi Nagahara.


British Journal of Pharmacology | 2003

A novel immunosuppressive agent FTY720 induced Akt dephosphorylation in leukemia cells

Yumiko Matsuoka; Yukitoshi Nagahara; Masahiko Ikekita; Takahisa Shinomiya

Our previous studies revealed that the immunosuppressive agent, FTY720, mainly induces mitochondria‐involved apoptosis in some types of cancer cells, since Bcl‐2 overexpression prevents the FTY720‐induction of apoptotic stimuli. Furthermore, FTY720 induces G0/G1 cell cycle arrest. The present study further examines the correlation between intracellular signaling kinases with FTY720‐induced mitochondria‐involved apoptosis. Human T cell leukemia Jurkat was exposed to FTY720. Dephosphorylation of Akt occurred in a time‐ and concentration‐dependent manner. FTY720 also induced Bad (Ser136) and ribosomal p70S6 kinase (p70S6k) (Thr389) dephosphorylation. FTY720‐induced Akt dephosphorylation was not because of Akt upstream phosphatidylinositol 3′‐kinase (PI 3‐kinase) pathway inhibition. FTY720 also induced Akt dephosphorylation in human B cell leukemia BALL‐1. BALL‐1 cells were resistant to FTY720‐induced apoptosis. Okadaic acid (OA) inhibited the FTY720‐induced dephosphorylation of Akt and p70S6k, suggesting that FTY720 promotes Ser/Thr protein phosphatase (PP) activity. OA partially inhibited FTY720‐induced caspase‐3 activation. PP2A or PP2A‐like phosphatase was temporarily activated in cells exposed to FTY720. In addition, FTY720 activated purified PP2A (ABC). Overall, the results suggest that FTY720 activated PP2A or PP2A‐like phosphatase and dephosphorylated Akt pathway factors resulting in the enhancement of apoptosis via mitochondria.


Journal of Immunology | 2000

Immunosuppressant FTY720 Induces Apoptosis by Direct Induction of Permeability Transition and Release of Cytochrome c from Mitochondria

Yukitoshi Nagahara; Masahiko Ikekita; Takahisa Shinomiya

FTY720 has immunosuppressive activity in experimental organ transplantation and shows a prompt and protracted decrease of blood T lymphocytes upon oral administration. The blood lymphocyte decrease in vivo was mainly a result of FTY720-induced apoptosis. However, this apoptotic mechanism is not well understood. We examined the mechanism of FTY720-induced apoptosis in lymphoma. Western blotting and fluorescent caspase-specific substrate revealed that caspase-3 is involved in FTY720-induced apoptosis, whereas caspase-1 is not. Apoptotic cell death was inhibited by the pan-caspase inhibitor, Z-VAD-FMK, suggesting that caspase activation is essential for FTY720-induced apoptosis. FTY720 reduced mitochondrial transmembrane potential and released cytochrome c from the mitochondria of intact cells as well as in a cell-free system even in the presence of Z-VAD-FMK. As these mitochondrial reactions occurred before caspase activation, we concluded that FTY720 directly influences mitochondrial functions. The inhibition of mitochondrial permeability transition by Bcl-2 overexpression or by chemical inhibitors prevented all apoptotic events occurring in intact cells and in a cell-free system. Moreover, using a cell-free system, FTY720 did not directly affect isolated nuclei or cytosol. These results indicate that FTY720 directly affects mitochondria and triggers permeability transition to induce further apoptotic events.


Immunopharmacology | 2000

Evidence that FTY720 induces T cell apoptosis in vivo.

Yukitoshi Nagahara; Shin Enosawa; Masahiko Ikekita; Seiichi Suzuki; Takahisa Shinomiya

The immunosuppressant FTY720 induces a drastic decrease in blood lymphocytes, especially T cells; a decrease which is assumed to be the immunosuppressive mechanism of this drug. FTY720 causes cell death in vitro in lymphocytes and leukemia cells. However, the deletion mechanism of blood lymphocytes in vivo remains unclear. We investigated whether administration of FTY720 induced lymphocyte apoptosis in blood circulation. A marked decrease in the number of blood lymphocytes was observed within an hour after a single oral administration of FTY720 at doses of 5-10 mg/kg in rats and mice. Experiments using fluorescein isothiocyanate (FITC)-Annexin V and APO-BRDU methods revealed that FTY720 induced blood lymphocyte apoptosis in a dose-dependent manner. On the other hand, lymphocyte homing to Peyers patches was proposed as the mechanism underlying the blood lymphocyte decrease at these doses. However, similar results were obtained when using aly/aly mice, which lack Peyers patches and lymph nodes. Thus, we concluded that apoptosis of blood lymphocytes was induced immediately after administration of FTY720, and the cells could be immediately scavenged by phagocytes or the reticuloendothelial system in addition to Peyers patches homing. We also concluded that T cells were highly sensitive to FTY720, which induced apoptosis in vivo.


British Journal of Pharmacology | 2002

T cell selective apoptosis by a novel immunosuppressant, FTY720, is closely regulated with Bcl‐2

Yukitoshi Nagahara; Masahiko Ikekita; Takahisa Shinomiya

A novel immunosuppressant FTY720 caused a significant decrease in peripheral T lymphocytes, but not in B lymphocytes upon oral administration. This decrease was mainly a result of FTY720‐induced apoptosis. In this study, we confirmed FTY720‐induced T cell selective apoptosis using lymphoma cell lines in vitro. Viability loss, DNA fragmentation, Annexin V binding, and caspases activation (caspase‐3, ‐8, and ‐9) were observed in Jurkat cells (T lymphoma cells), but not significantly in BALL‐1 cells (B lymphoma cells). These results indicated that FTY720 selectively induced apoptosis in T cell lymphoma to a greater extent than in B cell lymphoma, a finding that is similar to the result observed when FTY720 was treated with T lymphocytes and B lymphocytes in vitro. FTY720 released cytochrome c from mitochondria in Jurkat intact cells as well as from isolated Jurkat mitochondria directly, but not from mitochondria in BALL‐1 cells nor from isolated BALL‐1 mitochondria. BALL‐1 cells and B cells had more abundant mitochondria‐localized anti‐apoptotic protein Bcl‐2 than did Jurkat cells and T cells. FTY720‐induced apoptosis is inhibited by the overexpression of Bcl‐2, suggesting that the cellular Bcl‐2 level regulates the sensitivity to FTY720.


Japanese Journal of Cancer Research | 2001

Coordinate Involvement of Cell Cycle Arrest and Apoptosis Strengthen the Effect of FTY720

Yukitoshi Nagahara; Yumiko Matsuoka; Kaoru Saito; Masahiko Ikekita; Shigesada Higuchi; Takahisa Shinomiya

A novel reagent, FTY720 (2‐amino‐2‐[2‐(4‐octylphenyl)ethyl]‐l,3‐propanediol hydrochloride), has been shown to induce a significant decrease of lymphocytes and lymphoma cells and is expected to be a potent immunosuppressant and anti‐tumor drug. The decrease in lymphocytes and lymphoma cells is mainly the result of FTY720‐induced apoptosis. FTY720 directly affects mitochondria and induces cell death. Moreover, FTY720 activates protein phosphatase (PP) 2A and affects anti‐apo‐ptotic intracellular signal transduction proteins to attenuate the anti‐apoptotic effect. In this study, we examined the relationship between FTY720‐induced apoptosis and cell cycle regulation. FTY720 induced apoptosis significantly at the GO/G1 phase and caused GO/G1 cell cycle arrest of the human lymphoma cell lines HL‐60RG and Jurkat. Simultaneously, retinoblastoma protein (pRB) was dephosphorylated, suggesting that dephosphorylation of pRB was related to FTY720‐induced GO/G1 cell cycle arrest. Because this dephosphorylation was completely blocked by a specific PP1/ 2A inhibitor, okadaic acid, it appears that FTY720‐activated PP2A is essential for FTY720‐induced cell cycle arrest. FTY720‐induced apoptosis was inhibited by Bcl‐2 overexpression in Jurkat cells, but this did not prevent FTY720‐induced cell cycle arrest, suggesting that the mechanism of FTY720‐induced cell cycle arrest is independent of the mechanism of FTY720‐induced apoptosis. These two independent pathways strengthen the effect of FTY720.


International Immunopharmacology | 2014

5-Aminolevulinic acid combined with ferrous iron enhances the expression of heme oxygenase-1

Yoshiaki Nishio; Masayuki Fujino; Mingyi Zhao; Takuya Ishii; Masahiro Ishizuka; Hidenori Ito; Kiwamu Takahashi; Fuminori Abe; Motowo Nakajima; Tohru Tanaka; Shigeru Taketani; Yukitoshi Nagahara; Xiao-Kang Li

5-Aminolevulinic acid (5-ALA) is the naturally occurring metabolic precursor of heme. Heme negatively regulates the Maf recognition element (MARE) binding- and repressing-activity of the Bach1 transcription factor through its direct binding to Bach1. Heme oxygenase (HO)-1 is an inducible enzyme that catalyzes the rate-limiting step in the oxidative degradation of heme to free iron, biliverdin and carbon monoxide. These metabolites of heme protect against apoptosis, inflammation and oxidative stress. Monocytes and macrophages play a critical role in the initiation, maintenance and resolution of inflammation. Therefore, the regulation of inflammation in macrophages is an important target under various pathophysiological conditions. In order to address the question of what is responsible for the anti-inflammatory effects of 5-ALA, the induction of HO-1 expression by 5-ALA and sodium ferrous citrate (SFC) was examined in macrophage cell line (RAW264 cells). HO-1 expression induced by 5-ALA combined with SFC (5-ALA/SFC) was partially inhibited by MEK/ERK and p38 MAPK inhibitor. The NF-E2-related factor 2 (Nrf2) was activated and translocated from the cytosol to the nucleus in response to 5-ALA/SFC. Nrf2-specific siRNA reduced the HO-1 expression. In addition, 5-ALA/SFC increased the intracellular levels of heme in cells. The increased heme indicated that the inactivation of Bach1 by heme supports the upregulation of HO-1 expression. Taken together, our data suggest that the exposure of 5-ALA/SFC to RAW264 cells enhances the HO-1 expression via MAPK activation along with the negative regulation of Bach1.


Biochemical Pharmacology | 2008

Synthesis and pharmacological evaluation of the novel pseudo-symmetrical tamoxifen derivatives as anti-tumor agents

Isamu Shiina; Yoshiyuki Sano; Kenya Nakata; Takaaki Kikuchi; Akane Sasaki; Masahiko Ikekita; Yukitoshi Nagahara; Yoshimune Hasome; Takao Yamori; Kanami Yamazaki

Four pseudo-symmetrical tamoxifen derivatives, RID-B (13), RID-C (14), RID-D (15), and bis(dimethylaminophenetole) (16), were synthesized via the novel three-component coupling reaction, and the structure-activity relationships of these pseudo-symmetrical tamoxifen derivatives were examined. It was discovered that 13 and 16 strongly inhibit the viability of the HL-60 human acute promyelocytic leukemia cell line, whereas 14 possesses a medium activity against the same cell line and 15 has no effect on the cell viability. The global anti-tumor activity of 13-16 against a variety of human cancer cells was assessed using a panel of 39 human cancer cell lines (JFCR 39), and it was shown that RID-B (13) strongly inhibited the growth of several cancer cell lines at concentrations of less than 1 microM (at 0.38 microM for SF-539 [central nervous system], at 0.58 microM for HT-29 [colon], at 0.20 microM for DMS114 [lung], at 0.21 microM for LOX-IMVI [melanoma], and at 0.23 microM for MKN74 [stomach]).


Cancer Science | 2005

Phytosphingosine induced mitochondria-involved apoptosis.

Yukitoshi Nagahara; Takahisa Shinomiya; Sachiko Kuroda; Naoki Kaneko; Reiji Nishio; Masahiko Ikekita

Sphingolipids are putative intracellular signal mediators in cell differentiation, growth inhibition, and apoptosis. Sphingosine, sphinganine, and phytosphingosine are structural analogs of sphingolipids and are classified as long‐chain sphingoid bases. Sphingosine and sphinganine are known to play important roles in apoptosis. In the present study, we examined the phytosphingosine‐induced apoptosis mechanism, focusing on mitochondria in human T‐cell lymphoma Jurkat cells. Phytosphingosine significantly induced chromatin DNA fragmentation, which is a hallmark of apoptosis. Enzymatic activity measurements of caspases revealed that caspase‐3 and caspase‐9 are activated in phytosphingosine‐induced apoptosis, but there is little activation of caspase‐8 suggesting that phytosphingosine influences mitochondrial functions. In agreement with this hypothesis, a decrease in ΔΨm and the release of cytochrome c to the cytosol were observed upon phytosphingosine treatment. Furthermore, overexpression of mitochondria‐localized anti‐apoptotic protein Bcl‐2 prevented phytosphingosine apoptotic stimuli. Western blot assays revealed that phytosphingosine decreases phosphorylated Akt and p70S6k. Dephosphorylation of Akt was partially inhibited by protein phosphatase inhibitor OA and OA attenuated phytosphingosine‐induced apoptosis. Moreover, using a cell‐free system, phytosphingosine directly reduced ΔΨm. These results indicate that phytosphingosine perturbs mitochondria both directly and indirectly to induce apoptosis. (Cancer Sci 2005; 96: 83–92)


Cancer Science | 2008

Induction of mitochondria‐involved apoptosis in estrogen receptor‐negative cells by a novel tamoxifen derivative, ridaifen‐B

Yukitoshi Nagahara; Isamu Shiina; Kenya Nakata; Akane Sasaki; Tomomi Miyamoto; Masahiko Ikekita

Tamoxifen is an antagonist of estrogen receptor, which is used widely as an estrogen receptor‐positive breast cancer drug that blocks growth signals and provokes apoptosis. However, recent studies have revealed that tamoxifen induces apoptosis even in estrogen receptor‐negative cells. In the present study, we synthesized several tamoxifen derivatives to augment the apoptosis‐inducing effect of tamoxifen and evaluated the apoptosis‐inducing pathway. The estrogen receptor‐positive human leukemia cell line HL‐60 and estrogen receptor‐negative human leukemia cell line Jurkat were treated with tamoxifen and synthesized tamoxifen derivatives, and thereafter subjected to cell viability‐detection assays. Tamoxifen derivatives, as well as the lead compound tamoxifen, decreased the cell viability despite the expression of estrogen receptor. Among all of the synthesized tamoxifen derivatives, ridaifen‐B had more potent cancer cell‐damaging activity than tamoxifen. Ridaifen‐B fragmented Jurkat cell DNA and activated caspases, suggesting that the ridaifen‐B‐induced apoptosis pathway is estrogen receptor independent. Moreover, mitochondrial involvement during ridaifen‐B‐induced apoptosis was estimated. Ridaifen‐B significantly reduced mitochondrial membrane potential, and overexpression of Bcl‐2 inhibited ridaifen‐B‐induced apoptosis. These results suggest that the induction of apoptosis by ridaifen‐B, a novel tamoxifen derivative, is dependent on mitochondrial perturbation without estrogen receptor involvement. (Cancer Sci 2008; 99: 608–614)


Biochemical and Biophysical Research Communications | 2013

Novel tamoxifen derivative Ridaifen-B induces Bcl-2 independent autophagy without estrogen receptor involvement.

Yukitoshi Nagahara; Midori Takeyoshi; Seiya Sakemoto; Isamu Shiina; Kenya Nakata; Keiko Fujimori; Yanwen Wang; Eri Umeda; Chihiro Watanabe; Shoko Uetake; Takao Yamori; Shingo Dan; Yoji Yoshimi; Takahisa Shinomiya; Masahiko Ikekita

Autophagy is a self-proteolysis process in eukaryotic cells that results in the sequestering of intracellular proteins and organelles in autophagosomes. Activation of autophagy progress continued growth of some tumors, instead extensive autophagy induces cell death. In a previous study, we synthesized a novel tamoxifen derivative, Ridaifen (RID)-B. RID-B induced mitochondria-involved apoptosis even in estrogen receptor (ER)-negative cells. Since tamoxifen induces autophagy other than apoptosis, we treated ER-negative Jurkat cells with RID-B in the present study. RID-B treatment induced apoptosis and LC3 and lysosome colocalization, which results in the formation of autolysosomes. Western blotting revealed that LC3 was converted to LC3-I to LC3-II with RID-B treatment, suggesting that RID-B induced autophagy without ER involvement. Moreover, overexpression of the anti-apoptotic protein Bcl-2 suppressed the RID-B-induced cell death, but not the induction of autophagy. These results presumed that RID-B-induced autophagy is independent of Bcl-2, making RID-B-induced autophagy different from RID-B-induced apoptosis. Since Beclin 1 level is unchanged during RID-B treatment, RID-B induced autophagy pathway is Bcl-2/Beclin1 independent noncanonical pathway.

Collaboration


Dive into the Yukitoshi Nagahara's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Masahiko Ikekita

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar

Isamu Shiina

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar

Kenya Nakata

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Akane Sasaki

Tokyo University of Science

View shared research outputs
Top Co-Authors

Avatar

Taro Saito

Tokyo Denki University

View shared research outputs
Researchain Logo
Decentralizing Knowledge