Network


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

Hotspot


Dive into the research topics where Tomokazu Sumida is active.

Publication


Featured researches published by Tomokazu Sumida.


Cell | 2012

Complement C1q Activates Canonical Wnt Signaling and Promotes Aging-Related Phenotypes

Atsuhiko T. Naito; Tomokazu Sumida; Seitaro Nomura; Mei-Lan Liu; Tomoaki Higo; Akito Nakagawa; Katsuki Okada; Taku Sakai; Akihito Hashimoto; Yurina Hara; Ippei Shimizu; Weidong Zhu; Haruhiro Toko; Akemi Katada; Hiroshi Akazawa; Toru Oka; Jong-Kook Lee; Tohru Minamino; Toshio Nagai; Kenneth Walsh; Akira Kikuchi; Misako Matsumoto; Marina Botto; Ichiro Shiojima; Issei Komuro

Wnt signaling plays critical roles in development of various organs and pathogenesis of many diseases, and augmented Wnt signaling has recently been implicated in mammalian aging and aging-related phenotypes. We here report that complement C1q activates canonical Wnt signaling and promotes aging-associated decline in tissue regeneration. Serum C1q concentration is increased with aging, and Wnt signaling activity is augmented during aging in the serum and in multiple tissues of wild-type mice, but not in those of C1qa-deficient mice. C1q activates canonical Wnt signaling by binding to Frizzled receptors and subsequently inducing C1s-dependent cleavage of the ectodomain of Wnt coreceptor low-density lipoprotein receptor-related protein 6. Skeletal muscle regeneration in young mice is inhibited by exogenous C1q treatment, whereas aging-associated impairment of muscle regeneration is restored by C1s inhibition or C1qa gene disruption. Our findings therefore suggest the unexpected role of complement C1q in Wnt signal transduction and modulation of mammalian aging.


Circulation Research | 2010

Promotion of CHIP-Mediated p53 Degradation Protects the Heart From Ischemic Injury

Atsuhiko T. Naito; Sho Okada; Tohru Minamino; Koji Iwanaga; Mei-Lan Liu; Tomokazu Sumida; Seitaro Nomura; Naruhiko Sahara; Tatsuya Mizoroki; Akihiko Takashima; Hiroshi Akazawa; Toshio Nagai; Ichiro Shiojima; Issei Komuro

Rationale: The number of patients with coronary heart disease, including myocardial infarction, is increasing and novel therapeutic strategy is awaited. Tumor suppressor protein p53 accumulates in the myocardium after myocardial infarction, causes apoptosis of cardiomyocytes, and plays an important role in the progression into heart failure. Objectives: We investigated the molecular mechanisms of p53 accumulation in the heart after myocardial infarction and tested whether anti-p53 approach would be effective against myocardial infarction. Methods and Results: Through expression screening, we found that CHIP (carboxyl terminus of Hsp70-interacting protein) is an endogenous p53 antagonist in the heart. CHIP suppressed p53 level by ubiquitinating and inducing proteasomal degradation. CHIP transcription was downregulated after hypoxic stress and restoration of CHIP protein level prevented p53 accumulation after hypoxic stress. CHIP overexpression in vivo prevented p53 accumulation and cardiomyocyte apoptosis after myocardial infarction. Promotion of CHIP function by heat shock protein (Hsp)90 inhibitor, 17-allylamino-17-demethoxy geldanamycin (17-AAG), also prevented p53 accumulation and cardiomyocyte apoptosis both in vitro and in vivo. CHIP-mediated p53 degradation was at least one of the cardioprotective effects of 17-AAG. Conclusions: We found that downregulation of CHIP level by hypoxia was responsible for p53 accumulation in the heart after myocardial infarction. Decreasing the amount of p53 prevented myocardial apoptosis and ameliorated ventricular remodeling after myocardial infarction. We conclude that anti-p53 approach would be effective to treat myocardial infarction.


Nature Communications | 2015

Complement C1q-induced activation of β-catenin signalling causes hypertensive arterial remodelling.

Tomokazu Sumida; Atsuhiko T. Naito; Seitaro Nomura; Akito Nakagawa; Tomoaki Higo; Akihito Hashimoto; Katsuki Okada; Taku Sakai; Masamichi Ito; Toshihiro Yamaguchi; Toru Oka; Hiroshi Akazawa; Jong-Kook Lee; Tohru Minamino; Stefan Offermanns; Tetsuo Noda; Marina Botto; Yoshio Kobayashi; Hiroyuki Morita; Ichiro Manabe; Toshio Nagai; Ichiro Shiojima; Issei Komuro

Hypertension induces structural remodelling of arteries, which leads to arteriosclerosis and end-organ damage. Hyperplasia of vascular smooth muscle cells (VSMCs) and infiltration of immune cells are the hallmark of hypertensive arterial remodelling. However, the precise molecular mechanisms of arterial remodelling remain elusive. We have recently reported that complement C1q activates β-catenin signalling independent of Wnts. Here, we show a critical role of complement C1-induced activation of β-catenin signalling in hypertensive arterial remodelling. Activation of β-catenin and proliferation of VSMCs were observed after blood-pressure elevation, which were prevented by genetic and chemical inhibition of β-catenin signalling. Macrophage depletion and C1qa gene deletion attenuated the hypertension-induced β-catenin signalling, proliferation of VSMCs and pathological arterial remodelling. Our findings unveil the link between complement C1 and arterial remodelling and suggest that C1-induced activation of β-catenin signalling becomes a novel therapeutic target to prevent arteriosclerosis in patients with hypertension.


Scientific Reports | 2015

Angiotensin II receptor blockade promotes repair of skeletal muscle through down-regulation of aging-promoting C1q expression.

Chizuru Yabumoto; Hiroshi Akazawa; Rie Yamamoto; Masamichi Yano; Yoko Kudo-Sakamoto; Tomokazu Sumida; Takehiro Kamo; Hiroki Yagi; Yu Shimizu; Akiko Saga-Kamo; Atsuhiko T. Naito; Toru Oka; Jong-Kook Lee; Jun-ichi Suzuki; Yasushi Sakata; Etsuko Uejima; Issei Komuro

Disruption of angiotensin II type 1 (AT1) receptor prolonged life span in mice. Since aging-related decline in skeletal muscle function was retarded in Atgr1a−/− mice, we examined the role of AT1 receptor in muscle regeneration after injury. Administration of AT1 receptor blocker irbesartan increased the size of regenerating myofibers, decreased fibrosis, and enhanced functional muscle recovery after cryoinjury. We recently reported that complement C1q, secreted by macrophages, activated Wnt/β-catenin signaling and promoted aging-related decline in regenerative capacity of skeletal muscle. Notably, irbesartan induced M2 polarization of macrophages, but reduced C1q expression in cryoinjured muscles and in cultured macrophage cells. Irbesartan inhibited up-regulation of Axin2, a downstream gene of Wnt/β-catenin pathway, in cryoinjured muscles. In addition, topical administration of C1q reversed beneficial effects of irbesartan on skeletal muscle regeneration after injury. These results suggest that AT1 receptor blockade improves muscle repair and regeneration through down-regulation of the aging-promoting C1q-Wnt/β-catenin signaling pathway.


Circulation-heart Failure | 2015

Wnt/β-Catenin Signaling Contributes to Skeletal Myopathy in Heart Failure via Direct Interaction With Forkhead Box O

Katsuki Okada; Atsuhiko T. Naito; Tomoaki Higo; Akito Nakagawa; Masato Shibamoto; Taku Sakai; Akihito Hashimoto; Yuki Kuramoto; Tomokazu Sumida; Seitaro Nomura; Masamichi Ito; Toshihiro Yamaguchi; Toru Oka; Hiroshi Akazawa; Jong-Kook Lee; Sachio Morimoto; Yasushi Sakata; Ichiro Shiojima; Issei Komuro

Background—There are changes in the skeletal muscle of patients with chronic heart failure (CHF), such as volume reduction and fiber type shift toward fatigable type IIb fiber. Forkhead box O (FoxO) signaling plays a critical role in the development of skeletal myopathy in CHF, and functional interaction between FoxO and the Wnt signal mediator &bgr;-catenin was previously demonstrated. We have recently reported that serum of CHF model mice activates Wnt signaling more potently than serum of control mice and that complement C1q mediates this activation. We, therefore, hypothesized that C1q-induced activation of Wnt signaling plays a critical role in skeletal myopathy via the interaction with FoxO. Methods and Results—Fiber type shift toward fatigable fiber was observed in the skeletal muscle of dilated cardiomyopathy model mice, which was associated with activation of both Wnt and FoxO signaling. Wnt3a protein activated FoxO signaling and induced fiber type shift toward fatigable fiber in C2C12 cells. Wnt3a-induced fiber type shift was inhibited by suppression of FoxO1 activity, whereas Wnt3a-independent fiber type shift was observed by overexpression of constitutively active FoxO1. Serum of dilated cardiomyopathy mice activated both Wnt and FoxO signaling and induced fiber type shift toward fatigable fiber in C2C12 cells. Wnt inhibitor and C1-inhibitor attenuated FoxO activation and fiber type shift both in C2C12 cells and in the skeletal muscle of dilated cardiomyopathy mice. Conclusions—C1q-induced activation of Wnt signaling contributes to fiber type shift toward fatigable fiber in CHF. Wnt signaling may be a novel therapeutic target to prevent skeletal myopathy in CHF.


International Heart Journal | 2016

Generation of Induced Pluripotent Stem Cells From Patients With Duchenne Muscular Dystrophy and Their Induction to Cardiomyocytes.

Akihito Hashimoto; Atsuhiko T. Naito; Jong-Kook Lee; Rika Kitazume-Taneike; Masamichi Ito; Toshihiro Yamaguchi; Ryo Nakata; Tomokazu Sumida; Katsuki Okada; Akito Nakagawa; Tomoaki Higo; Yuki Kuramoto; Taku Sakai; Koji Tominaga; Takeshi Okinaga; Shigetoyo Kogaki; Keiichi Ozono; Shigeru Miyagawa; Yoshiki Sawa; Yasushi Sakata; Hiroyuki Morita; Akihiro Umezawa; Issei Komuro

Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene which encodes dystrophin protein. Dystrophin defect affects cardiac muscle as well as skeletal muscle. Cardiac dysfunction is observed in all patients with DMD over 18 years of age, but there is no curative treatment for DMD cardiomyopathy. To establish novel experimental platforms which reproduce the cardiac phenotype of DMD patients, here we established iPS cell lines from T lymphocytes donated from two DMD patients, with a protocol using Sendai virus vectors. We successfully conducted the differentiation of the DMD patient-specific iPS cells into beating cardiomyocytes. DMD patient-specific iPS cells and iPS cell-derived cardiomyocytes would be a useful in vitro experimental system with which to investigate DMD cardiomyopathy.


Scientific Reports | 2016

Activation of endothelial β-catenin signaling induces heart failure

Akito Nakagawa; Atsuhiko T. Naito; Tomokazu Sumida; Seitaro Nomura; Masato Shibamoto; Tomoaki Higo; Katsuki Okada; Taku Sakai; Akihito Hashimoto; Yuki Kuramoto; Toru Oka; Jong-Kook Lee; Mutsuo Harada; Kazutaka Ueda; Ichiro Shiojima; Florian P. Limbourg; Ralf H. Adams; Tetsuo Noda; Yasushi Sakata; Hiroshi Akazawa; Issei Komuro

Activation of β-catenin-dependent canonical Wnt signaling in endothelial cells plays a key role in angiogenesis during development and ischemic diseases, however, other roles of Wnt/β-catenin signaling in endothelial cells remain poorly understood. Here, we report that sustained activation of β-catenin signaling in endothelial cells causes cardiac dysfunction through suppressing neuregulin-ErbB pathway in the heart. Conditional gain-of-function mutation of β-catenin, which activates Wnt/β-catenin signaling in Bmx-positive arterial endothelial cells (Bmx/CA mice) led to progressive cardiac dysfunction and 100% mortality at 40 weeks after tamoxifen treatment. Electron microscopic analysis revealed dilatation of T-tubules and degeneration of mitochondria in cardiomyocytes of Bmx/CA mice, which are similar to the changes observed in mice with decreased neuregulin-ErbB signaling. Endothelial expression of Nrg1 and cardiac ErbB signaling were suppressed in Bmx/CA mice. The cardiac dysfunction of Bmx/CA mice was ameliorated by administration of recombinant neuregulin protein. These results collectively suggest that sustained activation of Wnt/β-catenin signaling in endothelial cells might be a cause of heart failure through suppressing neuregulin-ErbB signaling, and that the Wnt/β-catenin/NRG axis in cardiac endothelial cells might become a therapeutic target for heart failure.


Nature Communications | 2017

DNA single-strand break-induced DNA damage response causes heart failure

Tomoaki Higo; Atsuhiko T. Naito; Tomokazu Sumida; Masato Shibamoto; Katsuki Okada; Seitaro Nomura; Akito Nakagawa; Toshihiro Yamaguchi; Taku Sakai; Akihito Hashimoto; Yuki Kuramoto; Masamichi Ito; Shungo Hikoso; Hiroshi Akazawa; Jong-Kook Lee; Ichiro Shiojima; Peter J. McKinnon; Yasushi Sakata; Issei Komuro

The DNA damage response (DDR) plays a pivotal role in maintaining genome integrity. DNA damage and DDR activation are observed in the failing heart, however, the type of DNA damage and its role in the pathogenesis of heart failure remain elusive. Here we show the critical role of DNA single-strand break (SSB) in the pathogenesis of pressure overload-induced heart failure. Accumulation of unrepaired SSB is observed in cardiomyocytes of the failing heart. Unrepaired SSB activates DDR and increases the expression of inflammatory cytokines through NF-κB signalling. Pressure overload-induced heart failure is more severe in the mice lacking XRCC1, an essential protein for SSB repair, which is rescued by blocking DDR activation through genetic deletion of ATM, suggesting the causative role of SSB accumulation and DDR activation in the pathogenesis of heart failure. Prevention of SSB accumulation or persistent DDR activation may become a new therapeutic strategy against heart failure.


PLOS ONE | 2015

A Food-Derived Flavonoid Luteolin Protects against Angiotensin II-Induced Cardiac Remodeling

Atsuko Nakayama; Hiroyuki Morita; Tomoko Nakao; Toshihiro Yamaguchi; Tomokazu Sumida; Yuichi Ikeda; Hidetoshi Kumagai; Yoshihiro Motozawa; Tsukasa Takahashi; Atsushi Imaizumi; Tadashi Hashimoto; Ryozo Nagai; Issei Komuro

Oxidative stress has been implicated in cardiac remodeling (cardiac fibrosis and hypertrophy), which impairs cardiac function and metabolism; therefore, it is anticipated antioxidative compounds will have protective properties against cardiac remodeling. Luteolin (3’,4’,5,7-tetrahydroxyflavone), a widely distributed flavonoid found in many herbal extracts including celery, green pepper, perilla leaves and seeds, and chamomile, is a known to be a potent antioxidant and was previously demonstrated to exert an antifibrotic effect in the lungs and the liver. In this study, we clearly demonstrate that oral pretreatment with the higher-luteolin diet (0.035% (wt/wt)) protected against cardiac fibrosis and hypertrophy as well as a hyperoxidative state in Ang II-infused rats. In cardiac tissue, increased gene expression levels of TGFβ1, CTGF, Nox2, Nox4, ANP, and BNP induced by Ang II were restored by oral pretreatment of this high-luteolin diet. In cultured rat cardiac fibroblasts, H2O2-induced TGFβ1 expression and the phosphorylation of JNK were suppressed by luteolin pretreatment. In conclusion, food-derived luteolin has protective actions against Ang II-induced cardiac remodeling, which could be mediated through attenuation of oxidative stress.


Nature Communications | 2018

Cardiomyocyte gene programs encoding morphological and functional signatures in cardiac hypertrophy and failure

Seitaro Nomura; Masahiro Satoh; Takanori Fujita; Tomoaki Higo; Tomokazu Sumida; Toshiyuki Ko; Toshihiro Yamaguchi; Takashige Tobita; Atsuhiko T. Naito; Masamichi Ito; Kanna Fujita; Mutsuo Harada; Haruhiro Toko; Yoshio Kobayashi; Kaoru Ito; Eiki Takimoto; Hiroshi Akazawa; Hiroyuki Morita; Hiroyuki Aburatani; Issei Komuro

Pressure overload induces a transition from cardiac hypertrophy to heart failure, but its underlying mechanisms remain elusive. Here we reconstruct a trajectory of cardiomyocyte remodeling and clarify distinct cardiomyocyte gene programs encoding morphological and functional signatures in cardiac hypertrophy and failure, by integrating single-cardiomyocyte transcriptome with cell morphology, epigenomic state and heart function. During early hypertrophy, cardiomyocytes activate mitochondrial translation/metabolism genes, whose expression is correlated with cell size and linked to ERK1/2 and NRF1/2 transcriptional networks. Persistent overload leads to a bifurcation into adaptive and failing cardiomyocytes, and p53 signaling is specifically activated in late hypertrophy. Cardiomyocyte-specific p53 deletion shows that cardiomyocyte remodeling is initiated by p53-independent mitochondrial activation and morphological hypertrophy, followed by p53-dependent mitochondrial inhibition, morphological elongation, and heart failure gene program activation. Human single-cardiomyocyte analysis validates the conservation of the pathogenic transcriptional signatures. Collectively, cardiomyocyte identity is encoded in transcriptional programs that orchestrate morphological and functional phenotypes.The mechanisms underlying the transition from cardiac hypertrophy to heart failure following pressure overload are incompletely understood. Here the authors identify the gene programs encoding the morphological and functional characteristics of cardiomyocytes during the transition from early hypertrophy to heart failure via single-cell transcriptomics, establishing a key role for p53 signalling.

Collaboration


Dive into the Tomokazu Sumida'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

Ichiro Shiojima

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge