Network


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

Hotspot


Dive into the research topics where Takanori Numata is active.

Publication


Featured researches published by Takanori Numata.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2011

Accelerated epithelial cell senescence in IPF and the inhibitory role of SIRT6 in TGF-β-induced senescence of human bronchial epithelial cells

Shunsuke Minagawa; Jun Araya; Takanori Numata; Satoko Nojiri; Hiromichi Hara; Yoko Yumino; Makoto Kawaishi; Makoto Odaka; Toshiaki Morikawa; Stephen L. Nishimura; Katsutoshi Nakayama; Kazuyoshi Kuwano

Reepithelialization of remodeled air spaces with bronchial epithelial cells is a prominent pathological finding in idiopathic pulmonary fibrosis (IPF) and is implicated in IPF pathogenesis. Recent studies suggest that epithelial senescence is a risk factor for development of IPF, indicating such reepithelialization may be influenced by the acceleration of cellular senescence. Among the sirtuin (SIRT) family, SIRT6, a class III histone deacetylase, has been demonstrated to antagonize senescence. We evaluated the senescence of bronchiolization in association with SIRT6 expression in IPF lung. Senescence-associated β-galactosidase staining and immunohistochemical detection of p21 were performed to evaluate cellular senescence. As a model for transforming growth factor (TGF)-β-induced senescence of abnormal reepithelialization, we used primary human bronchial epithelial cells (HBEC). The changes of SIRT6, p21, and interleukin (IL)-1β expression levels in HBEC, as well as type I collagen expression levels in fibroblasts, were evaluated. In IPF lung samples, an increase in markers of senescence and SIRT6 expression was found in the bronchial epithelial cells lining cystically remodeled air spaces. We found that TGF-β induced senescence in primary HBEC by increasing p21 expression, and, whereas TGF-β also induced SIRT6, it was not sufficient to inhibit cellular senescence. However, overexpression of SIRT6 efficiently inhibited TGF-β-induced senescence via proteasomal degradation of p21. TGF-β-induced senescent HBEC secreted increased amounts of IL-1β, which was sufficient to induce myofibroblast differentiation in fibroblasts. These findings suggest that accelerated epithelial senescence plays a role in IPF pathogenesis through perpetuating abnormal epithelial-mesenchymal interactions, which can be antagonized by SIRT6.


OncoImmunology | 2012

Insufficient autophagy promotes bronchial epithelial cell senescence in chronic obstructive pulmonary disease

Satoko Fujii; Hiromichi Hara; Jun Araya; Naoki Takasaka; Jun-ichi Kojima; Saburo Ito; Shunsuke Minagawa; Yoko Yumino; Takeo Ishikawa; Takanori Numata; Makoto Kawaishi; Jun Hirano; Makoto Odaka; Toshiaki Morikawa; Stephen L. Nishimura; Katsutoshi Nakayama; Kazuyoshi Kuwano

Tobacco smoke-induced accelerated cell senescence has been implicated in the pathogenesis of chronic obstructive pulmonary disease (COPD). Cell senescence is accompanied by the accumulation of damaged cellular components suggesting that in COPD, inhibition of autophagy may contribute to cell senescence. Here we look at whether autophagy contributes to cigarette smoke extract (CSE) - induced cell senescence of primary human bronchial epithelial cells (HBEC), and further evaluate p62 and ubiquitinated protein levels in lung homogenates from COPD patients. We demonstrate that CSE transiently induces activation of autophagy in HBEC, followed by accelerated cell senescence and concomitant accumulation of p62 and ubiquitinated proteins. Autophagy inhibition further enhanced accumulations of p62 and ubiquitinated proteins, resulting in increased senescence and senescence-associated secretory phenotype (SASP) with interleukin (IL)-8 secretion. Conversely, autophagy activation by Torin1, a mammalian target of rapamycin (mTOR inhibitor), suppressed accumulations of p62 and ubiquitinated proteins and inhibits cell senescence. Despite increased baseline activity, autophagy induction in response to CSE was significantly decreased in HBEC from COPD patients. Increased accumulations of p62 and ubiquitinated proteins were detected in lung homogenates from COPD patients. Insufficient autophagic clearance of damaged proteins, including ubiquitinated proteins, is involved in accelerated cell senescence in COPD, suggesting a novel protective role for autophagy in the tobacco smoke-induced senescence-associated lung disease, COPD.


Journal of Immunology | 2014

Autophagy induction by SIRT6 through attenuation of insulin-like growth factor signaling is involved in the regulation of human bronchial epithelial cell senescence.

Naoki Takasaka; Jun Araya; Hiromichi Hara; Saburo Ito; Kenji Kobayashi; Yusuke Kurita; Hiroshi Wakui; Yutaka Yoshii; Yoko Yumino; Satoko Fujii; Shunsuke Minagawa; Chikako Tsurushige; Jun Kojima; Takanori Numata; Kenichiro Shimizu; Makoto Kawaishi; Yumi Kaneko; Noriki Kamiya; Jun Hirano; Makoto Odaka; Toshiaki Morikawa; Stephen L. Nishimura; Katsutoshi Nakayama; Kazuyoshi Kuwano

Cigarette smoke (CS)–induced cellular senescence has been implicated in the pathogenesis of chronic obstructive pulmonary disease, and SIRT6, a histone deacetylase, antagonizes this senescence, presumably through the attenuation of insulin-like growth factor (IGF)-Akt signaling. Autophagy controls cellular senescence by eliminating damaged cellular components and is negatively regulated by IGF-Akt signaling through the mammalian target of rapamycin (mTOR). SIRT1, a representative sirtuin family, has been demonstrated to activate autophagy, but a role for SIRT6 in autophagy activation has not been shown. Therefore, we sought to investigate the regulatory role for SIRT6 in autophagy activation during CS-induced cellular senescence. SIRT6 expression levels were modulated by cDNA and small interfering RNA transfection in human bronchial epithelial cells (HBECs). Senescence-associated β-galactosidase staining and Western blotting of p21 were performed to evaluate senescence. We demonstrated that SIRT6 expression levels were decreased in lung homogenates from chronic obstructive pulmonary disease patients, and SIRT6 expression levels correlated significantly with the percentage of forced expiratory volume in 1 s/forced vital capacity. CS extract (CSE) suppressed SIRT6 expression in HBECs. CSE-induced HBEC senescence was inhibited by SIRT6 overexpression, whereas SIRT6 knockdown and mutant SIRT6 (H133Y) without histone deacetylase activity enhanced HBEC senescence. SIRT6 overexpression induced autophagy via attenuation of IGF-Akt-mTOR signaling. Conversely, SIRT6 knockdown and overexpression of a mutant SIRT6 (H133Y) inhibited autophagy. Autophagy inhibition by knockdown of ATG5 and LC3B attenuated the antisenescent effect of SIRT6 overexpression. These results suggest that SIRT6 is involved in CSE-induced HBEC senescence via autophagy regulation, which can be attributed to attenuation of IGF-Akt-mTOR signaling.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2013

Mitochondrial fragmentation in cigarette smoke-induced bronchial epithelial cell senescence

Hiromichi Hara; Jun Araya; Saburo Ito; Kenji Kobayashi; Naoki Takasaka; Yutaka Yoshii; Hiroshi Wakui; Jun Kojima; Kenichiro Shimizu; Takanori Numata; Makoto Kawaishi; Noriki Kamiya; Makoto Odaka; Toshiaki Morikawa; Yumi Kaneko; Katsutoshi Nakayama; Kazuyoshi Kuwano

Mitochondria are dynamic organelles that continuously change their shape through fission and fusion. Disruption of mitochondrial dynamics is involved in disease pathology through excessive reactive oxygen species (ROS) production. Accelerated cellular senescence resulting from cigarette smoke exposure with excessive ROS production has been implicated in the pathogenesis of chronic obstructive pulmonary disease (COPD). Hence, we investigated the involvement of mitochondrial dynamics and ROS production in terms of cigarette smoke extract (CSE)-induced cellular senescence in human bronchial epithelial cells (HBEC). Mitochondrial morphology was examined by electron microscopy and fluorescence microscopy. Senescence-associated β-galactosidase staining and p21 Western blotting of primary HBEC were performed to evaluate cellular senescence. Mitochondrial-specific superoxide production was measured by MitoSOX staining. Mitochondrial fragmentation was induced by knockdown of mitochondrial fusion proteins (OPA1 or Mitofusins) by small-interfering RNA transfection. N-acetylcysteine and Mito-TEMPO were used as antioxidants. Mitochondria in bronchial epithelial cells were prone to be more fragmented in COPD lung tissues. CSE induced mitochondrial fragmentation and mitochondrial ROS production, which were responsible for acceleration of cellular senescence in HBEC. Mitochondrial fragmentation induced by knockdown of fusion proteins also increased mitochondrial ROS production and percentages of senescent cells. HBEC senescence and mitochondria fragmentation in response to CSE treatment were inhibited in the presence of antioxidants. CSE-induced mitochondrial fragmentation is involved in cellular senescence through the mechanism of mitochondrial ROS production. Hence, disruption of mitochondrial dynamics may be a part of the pathogenic sequence of COPD development.


Autophagy | 2015

PARK2-mediated mitophagy is involved in regulation of HBEC senescence in COPD pathogenesis.

Saburo Ito; Jun Araya; Yusuke Kurita; Kenji Kobayashi; Naoki Takasaka; Masahiro Yoshida; Hiromichi Hara; Shunsuke Minagawa; Hiroshi Wakui; Satoko Fujii; Jun Kojima; Kenichiro Shimizu; Takanori Numata; Makoto Kawaishi; Makoto Odaka; Toshiaki Morikawa; Toru Harada; Stephen L. Nishimura; Yumi Kaneko; Katsutoshi Nakayama; Kazuyoshi Kuwano

Cigarette smoke (CS)-induced mitochondrial damage with increased reactive oxygen species (ROS) production has been implicated in COPD pathogenesis by accelerating senescence. Mitophagy may play a pivotal role for removal of CS-induced damaged mitochondria, and the PINK1 (PTEN-induced putative kinase 1)-PARK2 pathway has been proposed as a crucial mechanism for mitophagic degradation. Therefore, we sought to investigate to determine if PINK1-PARK2-mediated mitophagy is involved in the regulation of CS extract (CSE)-induced cell senescence and in COPD pathogenesis. Mitochondrial damage, ROS production, and cell senescence were evaluated in primary human bronchial epithelial cells (HBEC). Mitophagy was assessed in BEAS-2B cells stably expressing EGFP-LC3B, using confocal microscopy to measure colocalization between TOMM20-stained mitochondria and EGFP-LC3B dots as a representation of autophagosome formation. To elucidate the involvement of PINK1 and PARK2 in mitophagy, knockdown and overexpression experiments were performed. PINK1 and PARK2 protein levels in lungs from patients were evaluated by means of lung homogenate and immunohistochemistry. We demonstrated that CSE-induced mitochondrial damage was accompanied by increased ROS production and HBEC senescence. CSE-induced mitophagy was inhibited by PINK1 and PARK2 knockdown, resulting in enhanced mitochondrial ROS production and cellular senescence in HBEC. Evaluation of protein levels demonstrated decreased PARK2 in COPD lungs compared with non-COPD lungs. These results suggest that PINK1-PARK2 pathway-mediated mitophagy plays a key regulatory role in CSE-induced mitochondrial ROS production and cellular senescence in HBEC. Reduced PARK2 expression levels in COPD lung suggest that insufficient mitophagy is a part of the pathogenic sequence of COPD.


American Journal of Respiratory Cell and Molecular Biology | 2012

Involvement of creatine kinase B in cigarette smoke-induced bronchial epithelial cell senescence.

Hiromichi Hara; Jun Araya; Naoki Takasaka; Satoko Fujii; Jun Kojima; Yoko Yumino; Kenichiro Shimizu; Takeo Ishikawa; Takanori Numata; Makoto Kawaishi; Keisuke Saito; Jun Hirano; Makoto Odaka; Toshiaki Morikawa; Hiroshi Hano; Katsutoshi Nakayama; Kazuyoshi Kuwano

Cigarette smoke induces damage to proteins and organelles by oxidative stress, resulting in accelerated epithelial cell senescence in the lung, which is implicated in chronic obstructive pulmonary disease (COPD) pathogenesis. Although the detailed molecular mechanisms are not fully understood, cellular energy status is one of the most crucial determinants for cell senescence. Creatine kinase (CK) is a constitutive enzyme, playing regulatory roles in energy homeostasis of cells. Among two isozymes, brain-type CK (CKB) is the predominant CK in lung tissue. In this study, we investigated the role of CKB in cigarette smoke extract (CSE)-induced cellular senescence in human bronchial epithelial cells (HBECs). Primary HBECs and Beas2B cells were used. Protein carbonylation was evaluated as a marker of oxidative protein damage. Cellular senescence was evaluated by senescence-associated β-galactosidase staining. CKB inhibition was examined by small interfering RNA and cyclocreatine. Secretion of IL-8, a hallmark of senescence-associated secretary phenotype, was measured by ELISA. CKB expression levels were reduced in HBECs from patients with COPD compared with that of HBECs from nonsmokers. CSE induced carbonylation of CKB and subsequently decreased CKB protein levels, which was reversed by a proteasome inhibitor. CKB inhibition alone induced cell senescence, and further enhanced CSE-induced cell senescence and IL-8 secretion. CSE-induced oxidation of CKB is a trigger for proteasomal degradation. Concomitant loss of enzymatic activity regulating energy homeostasis may lead to the acceleration of bronchial epithelial cell senescence, which is implicated in the pathogenesis of COPD.


Respiratory Research | 2016

Metformin attenuates lung fibrosis development via NOX4 suppression

Nahoko Sato; Naoki Takasaka; Masahiro Yoshida; Kazuya Tsubouchi; Shunsuke Minagawa; Jun Araya; Nayuta Saito; Yu Fujita; Yusuke Kurita; Kenji Kobayashi; Saburo Ito; Hiromichi Hara; Tsukasa Kadota; Haruhiko Yanagisawa; Mitsuo Hashimoto; Hirofumi Utsumi; Hiroshi Wakui; Jun Kojima; Takanori Numata; Yumi Kaneko; Makoto Odaka; Toshiaki Morikawa; Katsutoshi Nakayama; Hirotsugu Kohrogi; Kazuyoshi Kuwano

BackgroundAccumulation of profibrotic myofibroblasts in fibroblastic foci (FF) is a crucial process for development of fibrosis during idiopathic pulmonary fibrosis (IPF) pathogenesis, and transforming growth factor (TGF)-β plays a key regulatory role in myofibroblast differentiation. Reactive oxygen species (ROS) has been proposed to be involved in the mechanism for TGF-β-induced myofibroblast differentiation. Metformin is a biguanide antidiabetic medication and its pharmacological action is mediated through the activation of AMP-activated protein kinase (AMPK), which regulates not only energy homeostasis but also stress responses, including ROS. Therefore, we sought to investigate the inhibitory role of metformin in lung fibrosis development via modulating TGF-β signaling.MethodsTGF-β-induced myofibroblast differentiation in lung fibroblasts (LF) was used for in vitro models. The anti-fibrotic role of metfromin was examined in a bleomycin (BLM)-induced lung fibrosis model.ResultsWe found that TGF-β-induced myofibroblast differentiation was clearly inhibited by metformin treatment in LF. Metformin-mediated activation of AMPK was responsible for inhibiting TGF-β-induced NOX4 expression. NOX4 knockdown and N-acetylcysteine (NAC) treatment illustrated that NOX4-derived ROS generation was critical for TGF-β-induced SMAD phosphorylation and myofibroblast differentiation. BLM treatment induced development of lung fibrosis with concomitantly enhanced NOX4 expression and SMAD phosphorylation, which was efficiently inhibited by metformin. Increased NOX4 expression levels were also observed in FF of IPF lungs and LF isolated from IPF patients.ConclusionsThese findings suggest that metformin can be a promising anti-fibrotic modality of treatment for IPF affected by TGF-β.


Journal of Immunology | 2016

Involvement of PARK2-Mediated Mitophagy in Idiopathic Pulmonary Fibrosis Pathogenesis

Kenji Kobayashi; Jun Araya; Shunsuke Minagawa; Hiromichi Hara; Nayuta Saito; Tsukasa Kadota; Nahoko Sato; Masahiro Yoshida; Kazuya Tsubouchi; Yusuke Kurita; Saburo Ito; Yu Fujita; Naoki Takasaka; Hirofumi Utsumi; Haruhiko Yanagisawa; Mitsuo Hashimoto; Hiroshi Wakui; Jun Kojima; Kenichiro Shimizu; Takanori Numata; Makoto Kawaishi; Yumi Kaneko; Hisatoshi Asano; Makoto Yamashita; Makoto Odaka; Toshiaki Morikawa; Katsutoshi Nakayama; Kazuyoshi Kuwano

Fibroblastic foci, known to be the leading edge of fibrosis development in idiopathic pulmonary fibrosis (IPF), are composed of fibrogenic myofibroblasts. Autophagy has been implicated in the regulation of myofibroblast differentiation. Insufficient mitophagy, the mitochondria-selective autophagy, results in increased reactive oxygen species, which may modulate cell signaling pathways for myofibroblast differentiation. Therefore, we sought to investigate the regulatory role of mitophagy in myofibroblast differentiation as a part of IPF pathogenesis. Lung fibroblasts were used in in vitro experiments. Immunohistochemical evaluation in IPF lung tissues was performed. PARK2 was examined as a target molecule for mitophagy regulation, and a PARK2 knockout mouse was employed in a bleomycin-induced lung fibrosis model. We demonstrated that PARK2 knockdown-mediated mitophagy inhibition was involved in the mechanism for activation of the platelet-derived growth factor receptor (PDGFR)/PI3K/AKT signaling pathway accompanied by enhanced myofibroblast differentiation and proliferation, which were clearly inhibited by treatment with both antioxidants and AG1296, a PDGFR inhibitor. Mitophagy inhibition–mediated activation of PDGFR signaling was responsible for further autophagy suppression, suggesting the existence of a self-amplifying loop of mitophagy inhibition and PDGFR activation. IPF lung demonstrated reduced PARK2 with concomitantly increased PDGFR phosphorylation. Furthermore, bleomycin-induced lung fibrosis was enhanced in PARK2 knockout mice and subsequently inhibited by AG1296. These findings suggest that insufficient mitophagy-mediated PDGFR/PI3K/AKT activation, which is mainly attributed to reduced PARK2 expression, is a potent underlying mechanism for myofibroblast differentiation and proliferation in fibroblastic foci formation during IPF pathogenesis.


Journal of Immunology | 2011

Insulin-Dependent Phosphatidylinositol 3-Kinase/Akt and ERK Signaling Pathways Inhibit TLR3-Mediated Human Bronchial Epithelial Cell Apoptosis

Takanori Numata; Jun Araya; Satoko Fujii; Hiromichi Hara; Naoki Takasaka; Jun Kojima; Shunsuke Minagawa; Yoko Yumino; Makoto Kawaishi; Jun Hirano; Makoto Odaka; Toshiaki Morikawa; Stephen L. Nishimura; Katsutoshi Nakayama; Kazuyoshi Kuwano

TLR3, one of the TLRs involved in the recognition of infectious pathogens for innate and adaptive immunity, primarily recognizes viral-associated dsRNA. Recognition of dsRNA byproducts released from apoptotic and necrotic cells is a recently proposed mechanism for the amplification of toxicity, suggesting a pivotal participation of TLR3 in viral infection, as well as in lung diseases where apoptosis plays a critical role, such as asthma and chronic obstructive pulmonary disease. In addition to metabolic control, insulin signaling was postulated to be protective by inhibiting apoptosis. Therefore, we explored the role of insulin signaling in protecting against TLR3-mediated apoptosis of human bronchial epithelial cells. Significant TLR3-mediated apoptosis was induced by polyinosinic-polycytidylic acid, a dsRNA analog, via caspase-8–dependent mechanisms. However, insulin efficiently inhibited TLR3/ polyinosinic-polycytidylic acid-induced human bronchial epithelial cell apoptosis via PI3K/Akt and ERK pathways, at least in part, via upregulation of cellular FLIPs and through protein synthesis-independent mechanisms. These results indicate the significance of TLR3-mediated dsRNA-induced apoptosis in the pathogenesis of apoptosis-driven lung disease and provide evidence for a novel protective role of insulin.


Respiratory Research | 2013

Apoptosis inhibitor of macrophage (AIM) expression in alveolar macrophages in COPD

Jun-ichi Kojima; Jun Araya; Hiromichi Hara; Saburo Ito; Naoki Takasaka; Kenji Kobayashi; Satoko Fujii; Chikako Tsurushige; Takanori Numata; Takeo Ishikawa; Kenichiro Shimizu; Makoto Kawaishi; Keisuke Saito; Noriki Kamiya; Jun Hirano; Makoto Odaka; Toshiaki Morikawa; Hiroshi Hano; Satoko Arai; Toru Miyazaki; Yumi Kaneko; Katsutoshi Nakayama; Kazuyoshi Kuwano

BackgroundMarked accumulation of alveolar macrophages (AM) conferred by apoptosis resistance has been implicated in pathogenesis of chronic obstructive pulmonary disease (COPD). Apoptosis inhibitor of macrophage (AIM), has been shown to be produced by mature tissue macrophages and AIM demonstrates anti-apoptotic property against multiple apoptosis-inducing stimuli. Accordingly, we attempt to determine if AIM is expressed in AM and whether AIM is involved in the regulation of apoptosis in the setting of cigarette smoke extract (CSE) exposure.MethodsImmunohistochemical evaluations of AIM were performed. Immunostaining was assessed by counting total and positively staining AM numbers in each case (n = 5 in control, n = 5 in non-COPD smoker, n = 5 in COPD). AM were isolated from bronchoalveolar lavage fluid (BALF). The changes of AIM expression levels in response to CSE exposure in AM were evaluated. Knock-down of anti-apoptotic Bcl-xL was mediated by siRNA transfection. U937 monocyte-macrophage cell line was used to explore the anti-apoptotic properties of AIM.ResultsThe numbers of AM and AIM-positive AM were significantly increased in COPD lungs. AIM expression was demonstrated at both mRNA and protein levels in isolated AM, which was enhanced in response to CSE exposure. AIM significantly increased Bcl-xL expression levels in AM and Bcl-xL was involved in a part of anti-apoptotic mechanisms of AIM in U937 cells in the setting of CSE exposure.ConclusionsThese results suggest that AIM expression in association with cigarette smoking may be involved in accumulation of AM in COPD.

Collaboration


Dive into the Takanori Numata's collaboration.

Top Co-Authors

Avatar

Kazuyoshi Kuwano

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jun Araya

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Katsutoshi Nakayama

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hiromichi Hara

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Makoto Kawaishi

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jun Kojima

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Shunsuke Minagawa

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Yumi Kaneko

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Naoki Takasaka

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Saburo Ito

Jikei University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge