Network


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

Hotspot


Dive into the research topics where Masayuki Taniike is active.

Publication


Featured researches published by Masayuki Taniike.


Nature Medicine | 2007

The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress.

Atsuko Nakai; Osamu Yamaguchi; Toshihiro Takeda; Yoshiharu Higuchi; Shungo Hikoso; Masayuki Taniike; Shigemiki Omiya; Isamu Mizote; Yasushi Matsumura; Michio Asahi; Kazuhiko Nishida; Masatsugu Hori; Noboru Mizushima; Kinya Otsu

Autophagy, an evolutionarily conserved process for the bulk degradation of cytoplasmic components, serves as a cell survival mechanism in starving cells. Although altered autophagy has been observed in various heart diseases, including cardiac hypertrophy and heart failure, it remains unclear whether autophagy plays a beneficial or detrimental role in the heart. Here, we report that the cardiac-specific loss of autophagy causes cardiomyopathy in mice. In adult mice, temporally controlled cardiac-specific deficiency of Atg5 (autophagy-related 5), a protein required for autophagy, led to cardiac hypertrophy, left ventricular dilatation and contractile dysfunction, accompanied by increased levels of ubiquitination. Furthermore, Atg5-deficient hearts showed disorganized sarcomere structure and mitochondrial misalignment and aggregation. On the other hand, cardiac-specific deficiency of Atg5 early in cardiogenesis showed no such cardiac phenotypes under baseline conditions, but developed cardiac dysfunction and left ventricular dilatation one week after treatment with pressure overload. These results indicate that constitutive autophagy in the heart under baseline conditions is a homeostatic mechanism for maintaining cardiomyocyte size and global cardiac structure and function, and that upregulation of autophagy in failing hearts is an adaptive response for protecting cells from hemodynamic stress.


Molecular and Cellular Biology | 2004

p38α Mitogen-Activated Protein Kinase Plays a Critical Role in Cardiomyocyte Survival but Not in Cardiac Hypertrophic Growth in Response to Pressure Overload

Kazuhiko Nishida; Osaniu Yamaguchi; Shinichi Hirotani; Shungo Hikoso; Yoshiharu Higuchi; Tetsuya Watanabe; Toshihiro Takeda; Soh Osuka; Takashi Morita; Gen Kondoh; Yoshihiro Uno; Kazunori Kashiwase; Masayuki Taniike; Atsuko Nakai; Yasushi Matsumura; Jun-ichi Miyazaki; Tatsuhiko Sudo; Kenichi Hongo; Yoichiro Kusakari; Satoshi Kurihara; Kenneth R. Chien; Junji Takeda; Masatsugu Hori; Kinya Otsu

ABSTRACT The molecular mechanism for the transition from cardiac hypertrophy, an adaptive response to biomechanical stress, to heart failure is poorly understood. The mitogen-activated protein kinase p38α is a key component of stress response pathways in various types of cells. In this study, we attempted to explore the in vivo physiological functions of p38α in hearts. First, we generated mice with floxed p38α alleles and crossbred them with mice expressing the Cre recombinase under the control of the α-myosin heavy-chain promoter to obtain cardiac-specific p38α knockout mice. These cardiac-specific p38α knockout mice were born normally, developed to adulthood, were fertile, exhibited a normal life span, and displayed normal global cardiac structure and function. In response to pressure overload to the left ventricle, they developed significant levels of cardiac hypertrophy, as seen in controls, but also developed cardiac dysfunction and heart dilatation. This abnormal response to pressure overload was accompanied by massive cardiac fibrosis and the appearance of apoptotic cardiomyocytes. These results demonstrate that p38α plays a critical role in the cardiomyocyte survival pathway in response to pressure overload, while cardiac hypertrophic growth is unaffected despite its dramatic down-regulation.


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.


Biochemical and Biophysical Research Communications | 1992

Mitochondrial tRNAlle mutation in fatal cardiomyopathy

Masayuki Taniike; Hisao Fukushima; Itaru Yanagihara; Hiroko Tsukamoto; Junko Tanaka; Harutoshi Fujimura; Toshisaburo Nagai; T. Sano; K. Yamaoka; Koji Inui; Shintaro Okada

A patient with mitochondrial encephalomyopathy who died from progressive intractable cardiac failure at the age of 18 is reported. At the age of 4, he presented with short stature, but multiorgan disorders including deafness, focal glomerulosclerosis, epilepsy and dilated cardiomyopathy appeared later in his clinical course. Laboratory tests showed hyperlactatemia and hyperpyruvatemia. Histopathological findings demonstrated mitochondrial myopathy with ragged red fibers and focal cytochrome C oxidase-deficient fibers in skeletal and cardiac muscles. The activity of cytochrome C oxidase was 30% less than the control level in skeletal muscle. Sequencing of the entire mitochondrial tRNA genome revealed a novel point mutation in the tRNA(Ile) region (nt 4269). This A-to-G substitution was found in none of the 30 controls by screening using mispairing PCR and Ssp I digestion methods, suggesting that this new mutation was pathogenic in our case.


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.


The FASEB Journal | 2005

Presenilin 2 regulates the systolic function of heart by modulating Ca2+ signaling

Toshihiro Takeda; Michio Asahi; Osamu Yamaguchi; Shungo Hikoso; Hiroyuki Nakayama; Yoichiro Kusakari; Makoto Kawai; Kenichi Hongo; Yoshiharu Higuchi; Kazunori Kashiwase; Tetsuya Watanabe; Masayuki Taniike; Atsuko Nakai; Kazuhiko Nishida; Satoshi Kurihara; Dorit Donoviel; Alan Bernstein; Taisuke Tomita; Takeshi Iwatsubo; Masatsugu Hori; Kinya Otsu

Genetic studies of families with familial Alzheimers disease have implicated presenilin 2 (PS2) in the pathogenesis of this disease. PS2 is ubiquitously expressed in various tissues including hearts. In this study, we examined cardiac phenotypes of PS2 knockout (PS2KO) mice to elucidate a role of PS2 in hearts. PS2KO mice developed normally with no evidence of cardiac hypertrophy and fibrosis. Invasive hemodynamic analysis revealed that cardiac contractility in PS2KO mice increased compared with that in their littermate controls. A study of isolated papillary muscle showed that peak amplitudes of Ca2+ transients and peak tension were significantly higher in PS2KO mice than those in their littermate controls. PS2KO mouse hearts exhibited no change in expression of calcium regulatory proteins. Since it has been demonstrated that PS2 in brain interacts with sorcin, which serves as a modulator of cardiac ryanodine receptor (RyR2), we tested whether PS2 also interacts with RyR2. Immmunoprecipitation analysis showed that PS2, sorcin, and RyR2 interact with each other in HEK‐293 cells overexpressing these proteins or in mouse hearts. Immunohistochemistry of heart muscle indicated that PS2 colocalizes with RyR2 and sorcin at the Z‐lines. Elevated Ca2+ attenuated the association of RyR2 with PS2, whereas the association of sorcin with PS2 was enhanced. The enhanced Ca2+ transients and contractility in PS2KO mice were observed at low extracellular [Ca2+] but not at high levels of [Ca2+]. Taken together, our results suggest that PS2 plays an important role in cardiac excitation‐contraction coupling by interacting with RyR2.


Journal of the American College of Cardiology | 2008

Reduction in Hemoglobin–Oxygen Affinity Results in the Improvement of Exercise Capacity in Mice With Chronic Heart Failure

Tetsuya Watanabe; Toshihiro Takeda; Shigemiki Omiya; Shungo Hikoso; Osamu Yamaguchi; Yuko Nakano; Yoshiharu Higuchi; Atsuko Nakai; Yusuke Abe; Yayoi Aki-Jin; Masayuki Taniike; Isamu Mizote; Yasushi Matsumura; Takahiko Shimizu; Kazuhiko Nishida; Kiyohiro Imai; Masatsugu Hori; Takuji Shirasawa; Kinya Otsu

OBJECTIVES This study examined whether a reduction in hemoglobin-oxygen affinity improves exercise capacity in mice with heart failure. BACKGROUND Exercise intolerance is a major determinant of quality of life in patients with chronic heart failure. One of the major goals of the treatment for chronic heart failure is to improve quality of life. METHODS Four weeks after left coronary ligation, we transplanted bone marrow cells isolated from the transgenic mice expressing a hemoglobin variant with low oxygen affinity, Presbyterian, into the lethally irradiated mice with heart failure or administered a synthetic allosteric modifier of hemoglobin. The mice were then exercised on a treadmill. RESULTS Four weeks after the left coronary artery ligation, mice showed cardiac dysfunction and chamber dilation, which were characteristics of heart failure. The transplantation led to a reduction in hemoglobin-oxygen affinity and an increase in oxygen supply for skeletal muscle without changes in muscle properties. The transplanted mice showed improved running performance on a treadmill despite impaired cardiac contractility. Furthermore, administration of the synthetic allosteric modifier of hemoglobin showed a similar effect. CONCLUSIONS Allosteric modification of hemoglobin represents a therapeutic option for improving exercise capacity in patients with chronic heart failure. One mechanism of improvement in exercise capacity is enhanced oxygen delivery in the skeletal muscle.


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.

Collaboration


Dive into the Masayuki Taniike's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Takahiro Yoshimura

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge