Network


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

Hotspot


Dive into the research topics where Nahoko Sato is active.

Publication


Featured researches published by Nahoko Sato.


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.


Chest | 2013

Successful Treatment of Severe Amiodarone Pulmonary Toxicity With Polymyxin B-Immobilized Fiber Column Direct Hemoperfusion

Nahoko Sato; Keisuke Kojima; Yuko Horio; Eisuke Goto; Aiko Masunaga; Hidenori Ichiyasu; Hirotsugu Kohrogi

Amiodarone pulmonary toxicity (APT) is the most serious side effect of amiodarone. Although severe APT, such as ARDS, is rare, mortality of severe APT is high. Polymyxin B-immobilized fiber column direct hemoperfusion (PMX-DHP) is a medical device that reduces blood endotoxin levels in sepsis. Recent reports have shown that PMX-DHP improves oxygenation in patients with acute exacerbation of idiopathic pulmonary fibrosis and drug-induced severe interstitial pneumonia. Here, we present a case study of a patient with severe APT treated with PMX-DHP with complete recovery. The patient rapidly developed respiratory failure and required mechanical ventilation. Despite corticosteroid pulse therapy, no clinical improvement was noted. PMX-DHP was then started, and severe respiratory failure improved with reduction of serum levels of amiodarone and its metabolite monodesethylamiodarone. The patient was weaned from mechanical ventilation and has done well without recurrence. To our knowledge, this is the first reported case of PMX-DHP therapy for severe APT. We speculate that PMX-DHP could be a new treatment strategy for severe APT.


Respiratory Research | 2017

Pirfenidone inhibits myofibroblast differentiation and lung fibrosis development during insufficient mitophagy.

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

BackgroundPirfenidone (PFD) is an anti-fibrotic agent used to treat idiopathic pulmonary fibrosis (IPF), but its precise mechanism of action remains elusive. Accumulation of profibrotic myofibroblasts is a crucial process for fibrotic remodeling in IPF. Recent findings show participation of autophagy/mitophagy, part of the lysosomal degradation machinery, in IPF pathogenesis. Mitophagy has been implicated in myofibroblast differentiation through regulating mitochondrial reactive oxygen species (ROS)-mediated platelet-derived growth factor receptor (PDGFR) activation. In this study, the effect of PFD on autophagy/mitophagy activation in lung fibroblasts (LF) was evaluated, specifically the anti-fibrotic property of PFD for modulation of myofibroblast differentiation during insufficient mitophagy.MethodsTransforming growth factor-β (TGF-β)-induced or ATG5, ATG7, and PARK2 knockdown-mediated myofibroblast differentiation in LF were used for in vitro models. The anti-fibrotic role of PFD was examined in a bleomycin (BLM)-induced lung fibrosis model using PARK2 knockout (KO) mice.ResultsWe found that PFD induced autophagy/mitophagy activation via enhanced PARK2 expression, which was partly involved in the inhibition of myofibroblast differentiation in the presence of TGF-β. PFD inhibited the myofibroblast differentiation induced by PARK2 knockdown by reducing mitochondrial ROS and PDGFR-PI3K-Akt activation. BLM-treated PARK2 KO mice demonstrated augmentation of lung fibrosis and oxidative modifications compared to those of BLM-treated wild type mice, which were efficiently attenuated by PFD.ConclusionsThese results suggest that PFD induces PARK2-mediated mitophagy and also inhibits lung fibrosis development in the setting of insufficient mitophagy, which may at least partly explain the anti-fibrotic mechanisms of PFD for IPF treatment.


Autophagy | 2017

Azithromycin attenuates myofibroblast differentiation and lung fibrosis development through proteasomal degradation of NOX4

Kazuya Tsubouchi; Jun Araya; Shunsuke Minagawa; Hiromichi Hara; Akihiro Ichikawa; Nayuta Saito; Tsukasa Kadota; Nahoko Sato; Masahiro Yoshida; Yusuke Kurita; Kenji Kobayashi; Saburo Ito; Yu Fujita; Hirofumi Utsumi; Haruhiko Yanagisawa; Mitsuo Hashimoto; Hiroshi Wakui; Yutaka Yoshii; Takeo Ishikawa; Takanori Numata; Yumi Kaneko; Hisatoshi Asano; Makoto Yamashita; Makoto Odaka; Toshiaki Morikawa; Katsutoshi Nakayama; Yoichi Nakanishi; Kazuyoshi Kuwano

ABSTRACT Accumulation of profibrotic myofibroblasts is involved in the process of fibrosis development during idiopathic pulmonary fibrosis (IPF) pathogenesis. TGFB (transforming growth factor β) is one of the major profibrotic cytokines for myofibroblast differentiation and NOX4 (NADPH oxidase 4) has an essential role in TGFB-mediated cell signaling. Azithromycin (AZM), a second-generation antibacterial macrolide, has a pleiotropic effect on cellular processes including proteostasis. Hence, we hypothesized that AZM may regulate NOX4 levels by modulating proteostasis machineries, resulting in inhibition of TGFB-associated lung fibrosis development. Human lung fibroblasts (LF) were used to evaluate TGFB-induced myofibroblast differentiation. With respect to NOX4 regulation via proteostasis, assays for macroautophagy/autophagy, the unfolded protein response (UPR), and proteasome activity were performed. The potential anti-fibrotic property of AZM was examined by using bleomycin (BLM)-induced lung fibrosis mouse models. TGFB-induced NOX4 and myofibroblast differentiation were clearly inhibited by AZM treatment in LF. AZM-mediated NOX4 reduction was restored by treatment with MG132, a proteasome inhibitor. AZM inhibited autophagy and enhanced the UPR. Autophagy inhibition by AZM was linked to ubiquitination of NOX4 via increased protein levels of STUB1 (STIP1 homology and U-box containing protein 1), an E3 ubiquitin ligase. An increased UPR by AZM was associated with enhanced proteasome activity. AZM suppressed lung fibrosis development induced by BLM with concomitantly reduced NOX4 protein levels and enhanced proteasome activation. These results suggest that AZM suppresses NOX4 by promoting proteasomal degradation, resulting in inhibition of TGFB-induced myofibroblast differentiation and lung fibrosis development. AZM may be a candidate for the treatment of the fibrotic lung disease IPF.


Autophagy | 2018

PRKN-regulated mitophagy and cellular senescence during COPD pathogenesis.

Jun Araya; Kazuya Tsubouchi; Nahoko Sato; Saburo Ito; Shunsuke Minagawa; Hiromichi Hara; Yusuke Hosaka; Akihiro Ichikawa; Nayuta Saito; Tsukasa Kadota; Masahiro Yoshida; Yu Fujita; Hirofumi Utsumi; Kenji Kobayashi; Haruhiko Yanagisawa; Mitsuo Hashimoto; Hiroshi Wakui; Takeo Ishikawa; Takanori Numata; Yumi Kaneko; Hisatoshi Asano; Makoto Yamashita; Makoto Odaka; Toshiaki Morikawa; Stephen L. Nishimura; Katsutoshi Nakayama; Kazuyoshi Kuwano

ABSTRACT Cigarette smoke (CS)-induced accumulation of mitochondrial damage has been widely implicated in chronic obstructive pulmonary disease (COPD) pathogenesis. Mitophagy plays a crucial role in eliminating damaged mitochondria, and is governed by the PINK1 (PTEN induced putative protein kinase 1)-PRKN (parkin RBR E3 ubiquitin protein ligase) pathway. Although both increased PINK1 and reduced PRKN have been implicated in COPD pathogenesis in association with mitophagy, there are conflicting reports for the role of mitophagy in COPD progression. To clarify the involvement of PRKN-regulated mitophagy in COPD pathogenesis, prkn knockout (KO) mouse models were used. To illuminate how PINK1 and PRKN regulate mitophagy in relation to CS-induced mitochondrial damage and cellular senescence, overexpression and knockdown experiments were performed in airway epithelial cells (AEC). In comparison to wild-type mice, prkn KO mice demonstrated enhanced airway wall thickening with emphysematous changes following CS exposure. AEC in CS-exposed prkn KO mice showed accumulation of damaged mitochondria and increased oxidative modifications accompanied by accelerated cellular senescence. In vitro experiments showed PRKN overexpression was sufficient to induce mitophagy during CSE exposure even in the setting of reduced PINK1 protein levels, resulting in attenuation of mitochondrial ROS production and cellular senescence. Conversely PINK1 overexpression failed to recover impaired mitophagy caused by PRKN knockdown, indicating that PRKN protein levels can be the rate-limiting factor in PINK1-PRKN-mediated mitophagy during CSE exposure. These results suggest that PRKN levels may play a pivotal role in COPD pathogenesis by regulating mitophagy, suggesting that PRKN induction could mitigate the progression of COPD. Abbreviations: AD: Alzheimer disease; AEC: airway epithelial cells; BALF: bronchoalveolar lavage fluid; AKT: AKT serine/threonine kinase; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CDKN1A: cyclin dependent kinase inhibitor 1A; CDKN2A: cyclin dependent kinase inhibitor 2A; COPD: chronic obstructive pulmonary disease; CS: cigarette smoke; CSE: CS extract; CXCL1: C-X-C motif chemokine ligand 1; CXCL8: C-X-C motif chemokine ligand 8; HBEC: human bronchial epithelial cells; 4-HNE: 4-hydroxynonenal; IL: interleukin; KO: knockout; LF: lung fibroblasts; LPS: lipopolysaccharide; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MTOR: mechanistic target of rapamycin kinase; 8-OHdG: 8-hydroxy-2ʹ-deoxyguanosine; OPTN: optineurin; PRKN: parkin RBR E3 ubiquitin protein ligase; PCD: programmed cell death; PFD: pirfenidone; PIK3C: phosphatidylinositol-4:5-bisphosphate 3-kinase catalytic subunit; PINK1: PTEN induced putative kinase 1; PTEN: phosphatase and tensin homolog; RA: rheumatoid arthritis; ROS: reactive oxygen species; SA-GLB1/β-Gal: senescence-associated-galactosidase, beta 1; SASP: senescence-associated secretory phenotype; SNP: single nucleotide polymorphism; TNF: tumor necrosis factor.


Respiratory Care | 2016

Subinterlobular Pleural Location Is a Risk Factor for Pneumothorax After Bronchoscopy.

Haruka Chino; Motoyasu Iikura; Nayuta Saito; Nahoko Sato; Manabu Suzuki; Satoru Ishii; Eriko Morino; Go Naka; Jin Takasaki; Shinyu Izumi; Masayuki Hojo; Yuichiro Takeda; Haruhito Sugiyama

BACKGROUND: Pneumothorax is one of the most important complications after bronchoscopy. This study was conducted to determine the risk factors for post-bronchoscopy pneumothorax. METHODS: We retrospectively reviewed the medical records of 23 consecutive subjects who were diagnosed with iatrogenic pneumothorax after bronchoscopy between August 2010 and February 2014. Forty-six control subjects who did not develop pneumothorax after bronchoscopy were randomly selected. The factors affecting the occurrence of pneumothorax were determined by univariate and multivariate analyses. RESULTS: Among 991 patients who underwent bronchoscopy during the study period, 23 (2.3%) developed pneumothorax after bronchoscopy. Among these 23 subjects, 13 (57%) required chest tube drainage. Compared with the control group (46 randomly selected from 968 subjects who did not develop pneumothorax), the group that developed pneumothorax had a preponderance of women and had more target lesions located in the subpleural area (odds ratio [OR] 7.8, 95% CI 0.9–64), especially those that were close to the interlobular pleura (OR 5.1, 95% CI 1.6–16.1) and the left lung (OR 3.2, 95% CI 1.1–9.5). Multivariate analysis revealed that a subinterlobular pleural location of a lesion was a risk factor for pneumothorax (OR 4.8, 95% CI 1.1–20.4). CONCLUSIONS: Pneumothorax occurred significantly more frequently when bronchoscopy was performed for subinterlobular pleural lesions. Close attention and care should be taken during bronchoscopy, especially when target lesions are abutting the interlobular pleura.


European Respiratory Journal | 2017

PARK2-regulated mitophagy in COPD pathogenesis

Nahoko Sato; Kazuya Tsubouchi; Jun Araya; Shunsuke Minagawa; Hiromichi Hara; Nayuta Saito; Masahiro Yoshida; Yusuke Kurita; Kenji Kobayashi; Saburo Ito; Takeo Ishikawa; Takanori Numata; Yumi Kaneko; Katsutoshi Nakayama; Hirotsugu Kohrogi; Kazuyoshi Kuwano


European Respiratory Journal | 2017

Role of lamin B1 in COPD pathogenesis

Nayuta Saito; Jun Araya; Saburo Ito; Tsukasa Kadota; Masahiro Yoshida; Akihiro Ichikawa; Yusuke Kurita; Kazuya Tsubouchi; Nahoko Sato; Kenji Kobayashi; Shunsuke Minagawa; Hiromochi Hara; Hironori Kawamoto; Akihiko Ito; Hirofumi Utsumi; Haruhiko Yanagisama; Mitsuo Hashimoto; Hiroshi Wakui; Takeo Ishikawa; Takanori Numata; Yumi Kaneko; Katsutoshi Nakayama; Kazuyoshi Kuwano


European Respiratory Journal | 2017

Azithromycin-mediated CHIP regulation promotes proteasomal degradation of NOX4

Kazuya Tsubouchi; Jun Araya; Shunsuke Minagawa; Hiromichi Hara; Nayuta Saito; Akihiro Ichikawa; Nahoko Sato; Masahiro Yoshida; Yusuke Kurita; Takanori Numata; Yumi Kaneko; Katsutoshi Nakayama; Kazuyoshi Kuwano

Collaboration


Dive into the Nahoko Sato's collaboration.

Top Co-Authors

Avatar

Nayuta Saito

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

Kazuyoshi Kuwano

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Masahiro Yoshida

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

Takanori Numata

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

Katsutoshi Nakayama

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Kazuya Tsubouchi

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