Network


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

Hotspot


Dive into the research topics where Shunsuke Minagawa is active.

Publication


Featured researches published by Shunsuke Minagawa.


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.


Journal of Clinical Investigation | 2011

Mouse and human lung fibroblasts regulate dendritic cell trafficking, airway inflammation, and fibrosis through integrin αvβ8–mediated activation of TGF-β

Hideya Kitamura; Stephanie Cambier; Sangeeta Somanath; Tyren Barker; Shunsuke Minagawa; Jennifer A. Markovics; Amanda Goodsell; Louis F. Reichardt; David M. Jablons; Paul J. Wolters; Arthur Hill; James D. Marks; Jianlong Lou; Jean-Francois Pittet; Jack Gauldie; Jody L. Baron; Stephen L. Nishimura

The airway is a primary portal of entry for noxious environmental stimuli that can trigger airway remodeling, which contributes significantly to airway obstruction in chronic obstructive pulmonary disease (COPD) and chronic asthma. Important pathologic components of airway remodeling include fibrosis and abnormal innate and adaptive immune responses. The positioning of fibroblasts in interstitial spaces suggests that they could participate in both fibrosis and chemokine regulation of the trafficking of immune cells such as dendritic cells, which are crucial antigen-presenting cells. However, physiological evidence for this dual role for fibroblasts is lacking. Here, in two physiologically relevant models - conditional deletion in mouse fibroblasts of the TGF-β-activating integrin αvβ8 and neutralization of αvβ8 in human COPD fibroblasts - we have elucidated a mechanism whereby lung fibroblast chemokine secretion directs dendritic cell trafficking, in a manner that is critically dependent on αvβ8-mediated activation of TGF-β by fibroblasts. Our data therefore indicate that fibroblasts have a crucial role in regulating both fibrotic and immune responses in the lung.


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.


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.


Science Translational Medicine | 2014

Selective Targeting of TGF-β Activation to Treat Fibroinflammatory Airway Disease

Shunsuke Minagawa; Jianlong Lou; Robert Seed; Anthony Cormier; Shenping Wu; Yifan Cheng; Lynne A. Murray; Ping Tsui; Jane Connor; Ronald Herbst; Cedric Govaerts; Tyren Barker; Stephanie Cambier; Haruhiko Yanagisawa; Amanda Goodsell; Mitsuo Hashimoto; Oliver J. Brand; Ran Cheng; Royce Ma; Kate J. McKnelly; W. Wen; Arthur Hill; David M. Jablons; Paul J. Wolters; Hideya Kitamura; Jun Araya; Andrea J. Barczak; David J. Erle; Louis F. Reichardt; James D. Marks

Therapeutic targeting of an extended-closed conformation of the integrin αvβ8 inhibits TGF-β activation and ameliorates symptoms of experimental airway disease in mice. Breathing Freely Narrowing of the airways through accumulation of scar tissue and inflammation results from chronic injury in common diseases such as chronic obstructive pulmonary disease (COPD) and severe chronic asthma. Such airway narrowing causes the obstruction responsible for the breathlessness that these patients experience, and there are no available treatments that ameliorate fibroinflammatory airway narrowing. In a new study, Minagawa et al. engineered a monoclonal antibody that locks in a specific inactive conformation of a protein named integrin αvβ8. This protein is a crucial receptor required for activation of transforming growth factor–β, a central mediator of pathological inflammation and fibrosis. This antibody, when administered to mice engineered to express only human and not mouse αvβ8, reduced airway inflammation and fibrosis in response to a variety of injurious agents including cigarette smoke and allergens that are involved in the pathogenesis of COPD. Airway remodeling, caused by inflammation and fibrosis, is a major component of chronic obstructive pulmonary disease (COPD) and currently has no effective treatment. Transforming growth factor–β (TGF-β) has been widely implicated in the pathogenesis of airway remodeling in COPD. TGF-β is expressed in a latent form that requires activation. The integrin αvβ8 (encoded by the itgb8 gene) is a receptor for latent TGF-β and is essential for its activation. Expression of integrin αvβ8 is increased in airway fibroblasts in COPD and thus is an attractive therapeutic target for the treatment of airway remodeling in COPD. We demonstrate that an engineered optimized antibody to human αvβ8 (B5) inhibited TGF-β activation in transgenic mice expressing only human and not mouse ITGB8. The B5 engineered antibody blocked fibroinflammatory responses induced by tobacco smoke, cytokines, and allergens by inhibiting TGF-β activation. To clarify the mechanism of action of B5, we used hydrodynamic, mutational, and electron microscopic methods to demonstrate that αvβ8 predominantly adopts a constitutively active, extended-closed headpiece conformation. Epitope mapping and functional characterization of B5 revealed an allosteric mechanism of action due to locking-in of a low-affinity αvβ8 conformation. Collectively, these data demonstrate a new model for integrin function and present a strategy to selectively target the TGF-β pathway to treat fibroinflammatory airway diseases.


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.


Respiratory investigation | 2016

Cellular senescence and autophagy in the pathogenesis of chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF)

Kazuyoshi Kuwano; Jun Araya; Hiromichi Hara; Shunsuke Minagawa; Naoki Takasaka; Saburo Ito; Kenji Kobayashi; Katsutoshi Nakayama

Aging is associated with impairments in homeostasis. Although aging and senescence are not equivalent, the number of senescent cells increases with aging. Cellular senescence plays important roles in tissue repair or remodeling, as well as embryonic development. Autophagy is a process of lysosomal self-degradation that maintains a homeostatic balance between the synthesis, degradation, and recycling of cellular proteins. Autophagy diminishes with aging; additionally, accelerated aging can be attributed to reduced autophagy. Cellular senescence has been widely implicated in the pathogenesis of chronic obstructive pulmonary disease (COPD), a disease of accelerated lung aging, presumably by impairing cell repopulation and by aberrant cytokine secretion in the senescence-associated secretory phenotype. The possible participation of autophagy in the pathogenic sequence of COPD has been extensively explored. Although it has been reported that increased autophagy may induce epithelial cell death, an insufficient reserve of autophagy can induce cellular senescence in bronchial epithelial cells of COPD. Furthermore, advanced age is one of the most important risk factors for the development of idiopathic pulmonary fibrosis (IPF). Telomere shortening is found in blood leukocytes and alveolar epithelial cells from patients with IPF. Accelerated senescence of epithelial cells plays a role in IPF pathogenesis by perpetuating abnormal epithelial-mesenchymal interactions. Insufficient autophagy may be an underlying mechanism of accelerated epithelial cell senescence and myofibroblast differentiation in IPF. Herein, we review the molecular mechanisms of cellular senescence and autophagy and summarize the role of cellular senescence and autophagy in both COPD and IPF.


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.

Collaboration


Dive into the Shunsuke Minagawa's collaboration.

Top Co-Authors

Avatar

Jun Araya

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kazuyoshi Kuwano

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

Takanori Numata

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

Yumi Kaneko

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Saburo Ito

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hiroshi Wakui

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

Kenji Kobayashi

Jikei University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge