Network


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

Hotspot


Dive into the research topics where Ikuko Tsujimoto is active.

Publication


Featured researches published by Ikuko Tsujimoto.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Targeted deletion of apoptosis signal-regulating kinase 1 attenuates left ventricular remodeling

Osamu Yamaguchi; Yoshiharu Higuchi; Shinichi Hirotani; Kazunori Kashiwase; Hiroyuki Nakayama; Shungo Hikoso; Toshihiro Takeda; Tetsuya Watanabe; Michio Asahi; Masayuki Taniike; Yasushi Matsumura; Ikuko Tsujimoto; Kenichi Hongo; Yoichiro Kusakari; Satoshi Kurihara; Kazuhiko Nishida; Hidenori Ichijo; Masatsugu Hori; Kinya Otsu

Left ventricular remodeling that occurs after myocardial infarction (MI) and pressure overload is generally accepted as a determinant of the clinical course of heart failure. The molecular mechanism of this process, however, remains to be elucidated. Apoptosis signal-regulating kinase 1 (ASK1) is a mitogen-activated protein kinase kinase kinase that plays an important role in stress-induced apoptosis. We used ASK1 knockout mice (ASK-/-) to test the hypothesis that ASK1 is involved in development of left ventricular remodeling. ASK-/- hearts showed no morphological or histological defects. Echocardiography and cardiac catheterization revealed normal global structure and function. Left ventricular structural and functional remodeling were determined 4 weeks after coronary artery ligation or thoracic transverse aortic constriction (TAC). ASK-/- had significantly smaller increases in left ventricular end-diastolic and end-systolic ventricular dimensions and smaller decreases in fractional shortening in both experimental models compared with WT mice. The number of terminal deoxynucleotidyl transferase biotin-dUDP nick end-labeling-positive myocytes after MI or TAC was decreased in ASK-/- compared with that in WT mice. Overexpression of a constitutively active mutant of ASK1 induced apoptosis in isolated rat neonatal cardiomyocytes, whereas neonatal ASK-/- cardiomyocytes were resistant to H2O2-induced apoptosis. An in vitro kinase assay showed increased ASK1 activity in heart after MI or TAC in WT mice. Thus, ASK1 plays an important role in regulating left ventricular remodeling by promoting apoptosis.


Journal of Clinical Investigation | 2004

Cardiac-specific disruption of the c-raf-1 gene induces cardiac dysfunction and apoptosis

Osamu Yamaguchi; Tetsuya Watanabe; Kazuhiko Nishida; Kazunori Kashiwase; Yoshiharu Higuchi; Toshihiro Takeda; Shungo Hikoso; Shinichi Hirotani; Michio Asahi; Masayuki Taniike; Atsuko Nakai; Ikuko Tsujimoto; Yasushi Matsumura; Jun-ichi Miyazaki; Kenneth R. Chien; Atsushi Matsuzawa; Chiharu Sadamitsu; Hidenori Ichijo; Manuela Baccarini; Masatsugu Hori; Kinya Otsu

The Raf/MEK/extracellular signal-regulated kinase (ERK) signaling pathway regulates diverse cellular processes such as proliferation, differentiation, and apoptosis and is implicated as an important contributor to the pathogenesis of cardiac hypertrophy and heart failure. To examine the in vivo role of Raf-1 in the heart, we generated cardiac muscle-specific Raf-1-knockout (Raf CKO) mice with Cre-loxP-mediated recombination. The mice demonstrated left ventricular systolic dysfunction and heart dilatation without cardiac hypertrophy or lethality. The Raf CKO mice showed a significant increase in the number of apoptotic cardiomyocytes. The expression level and activation of MEK1/2 or ERK showed no difference, but the kinase activity of apoptosis signal-regulating kinase 1 (ASK1), JNK, or p38 increased significantly compared with that in controls. The ablation of ASK1 rescued heart dysfunction and dilatation as well as cardiac fibrosis. These results indicate that Raf-1 promotes cardiomyocyte survival through a MEK/ERK-independent mechanism.


Hypertension | 2005

The Antioxidant Edaravone Attenuates Pressure Overload–Induced Left Ventricular Hypertrophy

Ikuko Tsujimoto; Shungo Hikoso; Osamu Yamaguchi; Kazunori Kashiwase; Atsuko Nakai; Toshihiro Takeda; Tetsuya Watanabe; Masayuki Taniike; Yasushi Matsumura; Kazuhiko Nishida; Masatsugu Hori; Mikihiko Kogo; Kinya Otsu

The free radical scavenger 3-methyl-1-phenyl-2-pyrazolin-5-one (edaravone) is used to treat patients with ischemic brain damage. We and others reported previously that in vitro and in vivo reactive oxygen species (ROS) act as second messengers to develop cardiac hypertrophy. In this study, we used an in vivo murine model of pressure overload–induced cardiac hypertrophy to examine the effects of edaravone on left ventricular hypertrophy. The animals were subjected to the transverse thoracic aorta constriction, and edaravone (10 mg/kg) was infused intraperitoneally twice daily. Seven days after the operation, we observed a significant increase in ROS production in hearts, which was eliminated by the treatment with edaravone. Pressure-overloaded hearts showed a significant increase in left ventricular weight/body weight ratio and the expression level of atrial natriuretic factor mRNA, which were attenuated by edaravone. It also reduced perivascular and intermuscular fibrosis and inhibited pressure overload–induced activation of apoptosis signal-regulating kinase 1 (ASK1) and its downstream kinases of c-Jun N-terminal protein kinase and p38 mitogen-activated protein kinase. Edaravone attenuated the hypertrophic response even when the treatment was started after the onset of cardiac hypertrophic response. These findings indicate that edaravone significantly attenuates pressure overload–induced cardiac hypertrophy mediated through its antioxidative function and subsequent inhibition of ASK1 signaling pathway.


Circulation | 2008

Apoptosis Signal-Regulating Kinase 1/p38 Signaling Pathway Negatively Regulates Physiological Hypertrophy

Masayuki Taniike; Osamu Yamaguchi; Ikuko Tsujimoto; Shungo Hikoso; Toshihiro Takeda; Atsuko Nakai; Shigemiki Omiya; Isamu Mizote; Yuko Nakano; Yoshiharu Higuchi; Yasushi Matsumura; Kazuhiko Nishida; Hidenori Ichijo; Masatsugu Hori; Kinya Otsu

Background— Mechanical stress on the heart can lead to crucially different outcomes. Physiological stimuli such as exercise cause adaptive cardiac hypertrophy, characterized by a normal cardiac structure and normal or enhanced cardiac function. Pathological stimuli such as hypertension and aortic valvular stenosis cause maladaptive cardiac remodeling and ultimately heart failure. Apoptosis signal-regulating kinase 1 (ASK1) is known to be involved in pathological cardiac remodeling, but it has not been determined whether ASK1 pathways coordinate the signaling cascade leading to physiological type cardiac growth. Methods and Results— To evaluate the role of ASK1 in the physiological form of cardiac growth, mice lacking ASK1 (ASK1−/−) were exercised by swimming for 4 weeks. ASK1−/− mice showed exaggerated growth of the heart accompanied by typical characteristics of physiological hypertrophy. Their swimming-induced activation of Akt, a key molecule in the signaling cascade of physiological hypertrophy, increased more than that seen in wild-type controls. The activation of p38, a downstream kinase of ASK1, was suppressed selectively in the swimming-exercised ASK1−/− mice. Furthermore, the inhibition of ASK1 or p38 activity enhanced insulin-like growth factor 1–induced protein synthesis in rat neonatal ventricular cardiomyocytes, and the treatment with a specific inhibitor of p38 resulted in enhancement of Akt activation and suppression of protein phosphatase 2A activation. The cardiac-specific p38α-deficient mice developed an exacerbated form of cardiac hypertrophy in response to swimming exercise. Conclusions— These results indicate that the ASK1/p38 signaling pathway negatively regulates physiological hypertrophy.


Journal of Cell Science | 2013

The apical and basolateral secretion of Wnt11 and Wnt3a in polarized epithelial cells is regulated by different mechanisms.

Hideki Yamamoto; Chihiro Awada; Hideaki Hanaki; Hiroshi Sakane; Ikuko Tsujimoto; Yuko Takahashi; Toshifumi Takao; Akira Kikuchi

Summary Wnts are glycan- and lipid-modified morphogens that are important for cellular responses, but how Wnts are secreted in polarized epithelial cells remains unclear. Although Wntless (Wls) has been shown to interact with Wnts and support their secretion, the role of Wls in the sorting of Wnts to the final destination in polarized epithelial cells have not been clarified. Glycosylation was shown to be important for the sorting of some transmembrane and secreted proteins, but glycan profiles and their roles in the polarized secretion of Wnts has not yet been demonstrated. Here we show the apical and basolateral secretion of Wnts is regulated by different mechanisms. Wnt11 and Wnt3a were secreted apically and basolaterally, respectively, in polarized epithelial cells. Wls was localized to the basolateral membrane. Mass-spectrometric analyses revealed that Wnt11 is modified with complex/hybrid(Asn40)-, high-mannose(Asn90)- and high-mannose/hybrid(Asn300)-type glycans and that Wnt3a is modified with two high-mannose-type glycans (Asn87 and Asn298). Glycosylation processing at Asn40 and galectin-3 were required for the apical secretion of Wnt11, whereas clathrin and adaptor protein-1 were required for the basolateral secretion of Wnt3a. By the fusion of the Asn40 glycosylation site of Wnt11, Wnt3a was secreted apically. The recycling of Wls by AP-2 was necessary for the basolateral secretion of Wnt3a but not for the apical secretion of Wnt11. These results suggest that Wls has different roles in the polarized secretion of Wnt11 and Wnt3a and that glycosylation processing of Wnts decides their secretory routes.


Circulation | 2004

Pressure Overload Induces Cardiac Dysfunction and Dilation in Signal Transducer and Activator of Transcription 6–Deficient Mice

Shungo Hikoso; Osamu Yamaguchi; Yoshiharu Higuchi; Shinichi Hirotani; Toshihiro Takeda; Kazunori Kashiwase; Tetsuya Watanabe; Masayuki Taniike; Ikuko Tsujimoto; Michio Asahi; Yasushi Matsumura; Kazuhiko Nishida; Hiroshi Nakajima; Shizuo Akira; Masatsugu Hori; Kinya Otsu

Background—Signal transducer and activator of transcription (STAT) proteins constitute a family of transcription factors that mediate many cytokine-induced responses. STAT6 is activated by angiotensin II and in rat hypertrophied hearts and in human hearts with dilated cardiomyopathy. This suggests that STAT6 may be involved in the pathogenesis of cardiac hypertrophy and heart failure. For this study we used STAT6-deficient (STAT6−/−) mice to examine the in vivo role of STAT6. Methods and Results—STAT6−/− hearts showed no morphological, histological, or functional defects. We examined left ventricular structural and functional remodeling 1 week after thoracic transverse aortic constriction (TAC). Western blot and immunohistochemical analyses showed increased STAT6 activity after TAC in the heart of wild-type mice. STAT6−/− mice showed a significant increase in end-diastolic left ventricular internal dimension accompanied by impaired contractility compared with wild-type mice but no differences in hypertrophic parameters. The number of terminal deoxynucleotidyl transferase–mediated biotin dUTP nick-end labeling–positive myocytes after TAC had increased in STAT6−/− compared with wild-type mice. Prolonged induction of tumor necrosis factor-&agr; (TNF-&agr;) mRNA was observed in STAT6−/− hearts, whereas TNF-&agr; mRNA was only transiently induced in wild-type mice. Tristetraprolin was induced after TAC in wild-type mice but not in STAT6−/− mice. Tristetraprolin reporter assay with the use of isolated neonatal cardiomyocyte indicated that the promoter was significantly activated by endothelin-1 in wild-type but not in STAT6−/− cardiomyocytes. The lack of promoter activation by endothelin-1 in STAT6−/− cardiomyocytes was rescued by forced expression of STAT6. Conclusions—STAT6 plays a protective role against hemodynamic stress in hearts.


Scientific Reports | 2015

Wnt5a promotes cancer cell invasion and proliferation by receptor-mediated endocytosis-dependent and -independent mechanisms, respectively

Kensaku Shojima; Akira Sato; Hideaki Hanaki; Ikuko Tsujimoto; Masahiro Nakamura; Kazunari Hattori; Yuji Sato; Keiji Dohi; Michinari Hirata; Hideki Yamamoto; Akira Kikuchi

Wnt5a activates the Wnt/β-catenin-independent pathway and its overexpression is associated with tumor aggressiveness enhancing invasive activity. For this action, Wnt5a-induced receptor endocytosis with clathrin is required. Wnt5a expression was previously believed to be associated with cancer cell motility but not proliferation. Recently, it was reported that Wnt5a is also implicated in cancer cell proliferation, but the mechanism was not clear. In this study, we generated a neutralizing anti-Wnt5a monoclonal antibody (mAb5A16) to investigate the mechanism by which Wnt5a regulates cancer cell proliferation. Wnt5a stimulated both invasion and proliferation of certain types of cancer cells, including HeLaS3 cervical cancer cells and A549 lung cancer cells although Wnt5a promoted invasion but not proliferation in other cancer cells such as KKLS gastric cancer cells. mAb5A16 did not affect the binding of Wnt5a to its receptor, but it suppressed Wnt5a-induced receptor-mediated endocytosis. mAb5A16 inhibited invasion but not proliferation of HeLaS3 and A549 cells. Wnt5a activated Src family kinases (SFKs) and Wnt5a-dependent cancer cell proliferation was dependent on SFKs, yet blockade of receptor-mediated endocytosis did not affect cancer cell proliferation and SFK activity. These results suggest that Wnt5a promotes invasion and proliferation of certain types of cancer cells through receptor-mediated endocytosis-dependent and -independent mechanisms, respectively.


Journal of Molecular and Cellular Cardiology | 2004

Ca2+-sensitive tyrosine kinase Pyk2/CAK β-dependent signaling is essential for G-protein-coupled receptor agonist-induced hypertrophy

Shinichi Hirotani; Yoshiharu Higuchi; Kazuhiko Nishida; Hiroyuki Nakayama; Osamu Yamaguchi; Shungo Hikoso; Toshihiro Takeda; Kazunori Kashiwase; Tetsuya Watanabe; Michio Asahi; Masayuki Taniike; Ikuko Tsujimoto; Yasushi Matsumura; Terukatsu Sasaki; Masatsugu Hori; Kinya Otsu


Biochemical and Biophysical Research Communications | 2005

CaMKII activates ASK1 and NF-κB to induce cardiomyocyte hypertrophy

Kazunori Kashiwase; Yoshiharu Higuchi; Shinichi Hirotani; Osamu Yamaguchi; Shungo Hikoso; Toshihiro Takeda; Tetsuya Watanabe; Masayuki Taniike; Atsuko Nakai; Ikuko Tsujimoto; Yasushi Matsumura; Hikaru Ueno; Kazuhiko Nishida; Masatsugu Hori; Kinya Otsu


Southern African Journal of Anaesthesia and Analgesia | 2009

General anaesthesia with and without intubation for patients with Cornelia de Lange syndrome

Yoshinao Asahi; Ikuko Tsujimoto; Yusuke Kawai; Masahiro Sugimoto; Takahiro Suzuki; Shiro Omichi; Mikihiko Kogo; Junichiro Kotani

Collaboration


Dive into the Ikuko Tsujimoto's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge