Network


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

Hotspot


Dive into the research topics where Hitomi Hasegawa is active.

Publication


Featured researches published by Hitomi Hasegawa.


Cell Death and Disease | 2013

Jun dimerization protein 2 is a critical component of the Nrf2/MafK complex regulating the response to ROS homeostasis

S Tanigawa; Chien-Hung Lee; Chang-Shen Lin; Chia-Chen Ku; Hitomi Hasegawa; S Qin; A Kawahara; Y Korenori; K Miyamori; Michiya Noguchi; L H Lee; Yong Lin; C L Steve Lin; Yukio Nakamura; Chunyuan Jin; Naoto Yamaguchi; Richard Eckner; D-X Hou; Kazunari K. Yokoyama

Oxidative stress and reactive oxygen species (ROS) are associated with diseases such as cancer, cardiovascular complications, inflammation and neurodegeneration. Cellular defense systems must work constantly to control ROS levels and to prevent their accumulation. We report here that the Jun dimerization protein 2 (JDP2) has a critical role as a cofactor for transcription factors nuclear factor-erythroid 2-related factor 2 (Nrf2) and small Maf protein family K (MafK) in the regulation of the antioxidant-responsive element (ARE) and production of ROS. Chromatin immunoprecipitation–quantitative PCR (qPCR), electrophoresis mobility shift and ARE-driven reporter assays were carried out to examine the role of JDP2 in ROS production. JDP2 bound directly to the ARE core sequence, associated with Nrf2 and MafK (Nrf2–MafK) via basic leucine zipper domains, and increased DNA-binding activity of the Nrf2–MafK complex to the ARE and the transcription of ARE-dependent genes. In mouse embryonic fibroblasts from Jdp2-knockout (Jdp2 KO) mice, the coordinate transcriptional activation of several ARE-containing genes and the ability of Nrf2 to activate expression of target genes were impaired. Moreover, intracellular accumulation of ROS and increased thickness of the epidermis were detected in Jdp2 KO mice in response to oxidative stress-inducing reagents. These data suggest that JDP2 is required to protect against intracellular oxidation, ROS activation and DNA oxidation. qPCR demonstrated that several Nrf2 target genes such as heme oxygenase-1, glutamate–cysteine ligase catalytic and modifier subunits, the notch receptor ligand jagged 1 and NAD(P)H dehydrogenase quinone 1 are also dependent on JDP2 for full expression. Taken together, these results suggest that JDP2 is an integral component of the Nrf2–MafK complex and that it modulates antioxidant and detoxification programs by acting via the ARE.


Journal of Cell Science | 2013

Nuclear ErbB4 signaling through H3K9me3 is antagonized by EGFR-activated c-Src

Kenichi Ishibashi; Yasunori Fukumoto; Hitomi Hasegawa; Kohei Abe; Shoichi Kubota; Kazumasa Aoyama; Sho Kubota; Yuji Nakayama; Naoto Yamaguchi

Summary The ErbB family of receptor tyrosine kinases comprises four members: epidermal growth factor receptor (EGFR)/ErbB1, HER2/ErbB2, ErbB3 and ErbB4, and plays roles in signal transduction at the plasma membrane upon ligand stimulation. Stimulation with neuregulin-1 (NRG-1) cleaves ErbB4 and releases the ErbB4 intracellular domain (4ICD) that translocates into the nucleus to control gene expression. However, little is known about the regulation of 4ICD nuclear signaling through tyrosine phosphorylation. We show here that 4ICD nuclear signaling is antagonized by EGF-induced c-Src activation through EGFR. Generation of 4ICD by NRG-1 leads to increased levels of trimethylated histone H3 on lysine 9 (H3K9me3) in a manner dependent on the nuclear accumulation of 4ICD and its tyrosine kinase activity. Once EGF activates c-Src downstream of EGFR concomitantly with NRG-1-induced ErbB4 activation, c-Src associates with phospho-Tyr950 and phospho-Tyr1056 on 4ICD, thereby decreasing nuclear accumulation of 4ICD and inhibiting an increase of H3K9me3 levels. Moreover, 4ICD-induced transcriptional repression of the human telomerase reverse transcriptase (hTERT) is inhibited by EGF–EGFR–Src signaling. Thus, our findings reveal c-Src-mediated inhibitory regulation of ErbB4 nuclear signaling upon EGFR activation.


Oncogene | 2010

Suppression of cell-cycle progression by Jun dimerization protein-2 (JDP2) involves downregulation of cyclin-A2

Jianzhi Pan; Koji Nakade; Yu-Chang Huang; Zhu Zw; Masuzaki S; Hitomi Hasegawa; Takehide Murata; Yoshiki A; Naoto Yamaguchi; Lee Ch; Yang Wc; Eing-Mei Tsai; Yuuki Obata; Kazunari K. Yokoyama

We report here a novel role for Jun dimerization protein-2 (JDP2) as a regulator of the progression of normal cells through the cell cycle. To determine the role of JDP2 in vivo, we generated Jdp2-knockout (Jdp2KO) mice by targeting exon-1 to disrupt the site of initiation of transcription. The epidermal thickening of skin from the Jdp2KO mice after treatment with 12-O-tetradecanoylphorbol 13-acetate (TPA) proceeded more rapidly than that of control mice, and more proliferating cells were found at the epidermis. Fibroblasts derived from embryos of Jdp2KO mice proliferated faster and formed more colonies than fibroblasts from wild-type mice. JDP2 was recruited to the promoter of the gene for cyclin-A2 (ccna2) at the AP-1 site. Cells lacking Jdp2 had elevated levels of cyclin-A2 mRNA. Furthermore, reintroduction of JDP2 resulted in the repression of transcription of ccna2 and of cell-cycle progression. Thus, transcription of the gene for cyclin-A2 appears to be a direct target of JDP2 in the suppression of cell proliferation.We report here a novel role for Jun dimerization protein-2 (JDP2) as a regulator of the progression of normal cells through the cell cycle. To determine the role of JDP2 in vivo, we generated Jdp2-knockout (Jdp2KO) mice by targeting exon-1 to disrupt the site of initiation of transcription. The epidermal thickening of skin from the Jdp2KO mice after treatment with 12-O-tetradecanoylphorbol 13-acetate (TPA) proceeded more rapidly than that of control mice, and more proliferating cells were found at the epidermis. Fibroblasts derived from embryos of Jdp2KO mice proliferated faster and formed more colonies than fibroblasts from wild-type mice. JDP2 was recruited to the promoter of the gene for cyclin-A2 (ccna2) at the AP-1 site. Cells lacking Jdp2 had elevated levels of cyclin-A2 mRNA. Furthermore, reintroduction of JDP2 resulted in the repression of transcription of ccna2 and of cell-cycle progression. Thus, transcription of the gene for cyclin-A2 appears to be a direct target of JDP2 in the suppression of cell proliferation.


PLOS ONE | 2014

Cdk1-mediated phosphorylation of human ATF7 at Thr-51 and Thr-53 promotes cell-cycle progression into M phase.

Hitomi Hasegawa; Kenichi Ishibashi; Shoichi Kubota; Chihiro Yamaguchi; Ryuzaburo Yuki; Haruna Nakajo; Richard Eckner; Noritaka Yamaguchi; Kazunari K. Yokoyama; Naoto Yamaguchi

Activating transcription factor 2 (ATF2) and its homolog ATF7 are phosphorylated at Thr-69/Thr-71 and at Thr-51/Thr-53, respectively, by stress-activated MAPKs regulating their transcriptional functions in G1 and S phases. However, little is known about the role of ATF2 and ATF7 in G2/M phase. Here, we show that Cdk1-cyclin B1 phosphorylates ATF2 at Thr-69/Thr-71 and ATF7 at Thr-51/Thr-53 from early prophase to anaphase in the absence of any stress stimulation. Knockdown of ATF2 or ATF7 decreases the rate of cell proliferation and the number of cells in M-phase. In particular, the knockdown of ATF7 severely inhibits cell proliferation and G2/M progression. The inducible expression of a mitotically nonphosphorylatable version of ATF7 inhibits G2/M progression despite the presence of endogenous ATF7. We also show that mitotic phosphorylation of ATF7 promotes the activation of Aurora kinases, which are key enzymes for early mitotic events. These results suggest that the Cdk1-mediated phosphorylation of ATF7 facilitates G2/M progression, at least in part, by enabling Aurora signaling.


BioMed Research International | 2011

Jun dimerization protein 2 controls senescence and differentiation via regulating histone modification.

Yu-Chang Huang; Hitomi Hasegawa; Shin-Wei Wang; Chia-Chen Ku; Ying-Chu Lin; Shyh-Shin Chiou; Ming-Feng Hou; Deng-Chyang Wu; Eing-Mei Tsai; Shigeo Saito; Naoto Yamaguchi; Kazunari K. Yokoyama

Transcription factor, Jun dimerization protein 2 (JDP2), binds directly to histones and DNAs and then inhibits the p300-mediated acetylation both of core histones and of reconstituted nucleosomes that contain JDP2 recognition DNA sequences. JDP2 plays a key role as a repressor of adipocyte differentiation by regulation of the expression of the gene C/EBPδ via inhibition of histone acetylation. Moreover, JDP2-deficient mouse embryonic fibroblasts (JDP2−/− MEFs) are resistant to replicative senescence. JDP2 inhibits the recruitment of polycomb repressive complexes (PRC1 and PRC2) to the promoter of the gene encoding p16Ink4a, resulting from the inhibition of methylation of lysine 27 of histone H3 (H3K27). Therefore, it seems that chromatin-remodeling factors, including the PRC complex controlled by JDP2, may be important players in the senescence program. The novel mechanisms that underline the action of JDP2 in inducing cellular senescence and suppressing adipocyte differentiation are reviewed.


Biochemical and Biophysical Research Communications | 2014

Lyn tyrosine kinase promotes silencing of ATM-dependent checkpoint signaling during recovery from DNA double-strand breaks

Yasunori Fukumoto; Kazumasa Kuki; Mariko Morii; Takahito Miura; Takuya Honda; Kenichi Ishibashi; Hitomi Hasegawa; Sho Kubota; Yudai Ide; Noritaka Yamaguchi; Yuji Nakayama; Naoto Yamaguchi

DNA damage activates the DNA damage checkpoint and the DNA repair machinery. After initial activation of DNA damage responses, cells recover to their original states through completion of DNA repair and termination of checkpoint signaling. Currently, little is known about the process by which cells recover from the DNA damage checkpoint, a process called checkpoint recovery. Here, we show that Src family kinases promote inactivation of ataxia telangiectasia mutated (ATM)-dependent checkpoint signaling during recovery from DNA double-strand breaks. Inhibition of Src activity increased ATM-dependent phosphorylation of Chk2 and Kap1. Src inhibition increased ATM signaling both in G2 phase and during asynchronous growth. shRNA knockdown of Lyn increased ATM signaling. Src-dependent nuclear tyrosine phosphorylation suppressed ATM-mediated Kap1 phosphorylation. These results suggest that Src family kinases are involved in upstream signaling that leads to inactivation of the ATM-dependent DNA damage checkpoint.


Journal of Cellular Biochemistry | 2015

Overexpression of zinc-finger protein 777 (ZNF777) inhibits proliferation at low cell density through down-regulation of FAM129A.

Ryuzaburo Yuki; Kazumasa Aoyama; Sho Kubota; Noritaka Yamaguchi; Shoichi Kubota; Hitomi Hasegawa; Mariko Morii; Xiayu Huang; Kang Liu; Roy Williams; Michiko N. Fukuda; Naoto Yamaguchi

Krüppel‐associated box‐containing zinc finger proteins (KRAB‐ZFPs) regulate a wide range of cellular processes. KRAB‐ZFPs have a KRAB domain, which binds to transcriptional corepressors, and a zinc finger domain, which binds to DNA to activate or repress gene transcription. Here, we characterize ZNF777, a member of KRAB‐ZFPs. We show that ZNF777 localizes to the nucleus and inducible overexpression of ZNF777 inhibits cell proliferation in a manner dependent on its zinc finger domain but independent of its KRAB domain. Intriguingly, ZNF777 overexpression drastically inhibits cell proliferation at low cell density but slightly inhibits cell proliferation at high cell density. Furthermore, ZNF777 overexpression decreases the mRNA level of FAM129A irrespective of cell density. Importantly, the protein level of FAM129A strongly decreases at low cell density, but at high cell density the protein level of FAM129A does not decrease to that observed at low cell density. ZNF777‐mediated inhibition of cell proliferation is attenuated by overexpression of FAM129A at low cell density. Furthermore, ZNF777‐mediated down‐regulation of FAM129A induces moderate levels of the cyclin‐dependent kinase inhibitor p21. These results suggest that ZNF777 overexpression inhibits cell proliferation at low cell density and that p21 induction by ZNF777‐mediated down‐regulation of FAM129A plays a role in inhibition of cell proliferation. J. Cell. Biochem. 116: 954–968, 2015.


Cancers | 2013

Control of Oxidative Stress and Generation of Induced Pluripotent Stem Cell-like Cells by Jun Dimerization Protein 2

Shyh-Shin Chiou; Sophie Sheng-Wen Wang; Deng-Chyang Wu; Ying-Chu Lin; Li-Pin Kao; Kung-Kai Kuo; Chun-Chieh Wu; Chee-Yin Chai; Cheng-Lung Steve Lin; Cheng-Yi Lee; Yu-Mei Liao; Kenly Wuputra; Ya‐Han Yang; Shin-Wei Wang; Chia-Chen Ku; Yukio Nakamura; Shigeo Saito; Hitomi Hasegawa; Naoto Yamaguchi; Hiroyuki Miyoshi; Chang-Sheng Lin; Richard Eckner; Kazunari K. Yokoyama

We report here that the Jun dimerization protein 2 (JDP2) plays a critical role as a cofactor for the transcription factors nuclear factor-erythroid 2-related factor 2 (Nrf2) and MafK in the regulation of the antioxidants and production of reactive oxygen species (ROS). JDP2 associates with Nrf2 and MafK (Nrf2-MafK) to increase the transcription of antioxidant response element-dependent genes. Oxidative-stress-inducing reagent led to an increase in the intracellular accumulation of ROS and cell proliferation in Jdp2 knock-out mouse embryonic fibroblasts. In Jdp2-Cre mice mated with reporter mice, the expression of JDP2 was restricted to granule cells in the brain cerebellum. The induced pluripotent stem cells (iPSC)-like cells were generated from DAOY medulloblastoma cell by introduction of JDP2, and the defined factor OCT4. iPSC-like cells expressed stem cell-like characteristics including alkaline phosphatase activity and some stem cell markers. However, such iPSC-like cells also proliferated rapidly, became neoplastic, and potentiated cell malignancy at a later stage in SCID mice. This study suggests that medulloblastoma cells can be reprogrammed successfully by JDP2 and OCT4 to become iPSC-like cells. These cells will be helpful for studying the generation of cancer stem cells and ROS homeostasis.


Proceedings of the National Academy of Sciences of the United States of America | 1994

Structure, function, and evolution of mouse TL genes, nonclassical class I genes of the major histocompatibility complex.

Yuichi Obata; Yoko Satta; Kazuo Moriwaki; Toshihiko Shiroishi; Hitomi Hasegawa; Toshitada Takahashi; Naoyuki Takahata


Nature Precedings | 2009

Suppression of cell cycle progression by Jun dimerization protein (JDP2) involves down-regulation of cyclin A2

Jianzhi Pan; Koji Nakade; Satoko Masuzaki; Hitomi Hasegawa; Yu-Chang Huang; Takehide Murata; Atsushi Yoshiki; Naoto Yamaguchi; Yuichi Obata; Kazunari K. Yokoyama

Collaboration


Dive into the Hitomi Hasegawa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chia-Chen Ku

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar

Kazunari K. Yokoyama

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar

Yu-Chang Huang

Kaohsiung Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eing-Mei Tsai

Kaohsiung Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge