Network


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

Hotspot


Dive into the research topics where Kazutoshi Mawatari is active.

Publication


Featured researches published by Kazutoshi Mawatari.


Jacc-cardiovascular Imaging | 2011

Pioglitazone Attenuates Atherosclerotic Plaque Inflammation in Patients With Impaired Glucose Tolerance or Diabetes: A Prospective, Randomized, Comparator-Controlled Study Using Serial FDG PET/CT Imaging Study of Carotid Artery and Ascending Aorta

Minori Mizoguchi; Nobuhiro Tahara; Atsuko Tahara; Yoshikazu Nitta; Norihiro Kodama; Toyoharu Oba; Kazutoshi Mawatari; Hideo Yasukawa; Hayato Kaida; Masatoshi Ishibashi; Naofumi Hayabuchi; Haruhito Harada; Hisao Ikeda; Sho-ichi Yamagishi; Tsutomu Imaizumi

OBJECTIVES The aim of this study was to compare the effect of pioglitazone, an insulin sensitizer, with glimepiride, an insulin secretagogue, on atherosclerotic plaque inflammation by using serial (18)F-fluorodeoxyglucose positron emission tomography (FDG-PET) imaging. BACKGROUND Atherosclerosis is intrinsically an inflammatory disease. Although hyperglycemia is associated with an increased risk of atherosclerotic cardiovascular disease, there are no clinical data to show the preference of any specific oral hypoglycemic agents to prevent atherosclerotic plaque inflammation. METHODS A total of 56 impaired glucose tolerant or diabetic patients with carotid atherosclerosis underwent a complete history, determinations of blood chemistries, anthropometric variables, and FDG-PET. They were randomly assigned to receive either pioglitazone (15 to 30 mg) or glimepiride (0.5 to 4.0 mg) for 4 months with titration to optimal dosage. Effects of the drugs on atherosclerotic plaque inflammation were evaluated by FDG-PET at study completion. Plaque inflammation was measured by blood-normalized standardized uptake value, known as a target-to-background ratio. RESULTS The study was completed in 31 pioglitazone-treated patients and 21 glimepiride-treated patients. Although both treatments reduced fasting plasma glucose and hemoglobin A1c values comparably, pioglitazone, but not glimepiride, decreased atherosclerotic plaque inflammation. Compared with glimepiride, pioglitazone significantly increased high-density lipoprotein cholesterol level. High-sensitivity C-reactive protein was decreased by pioglitazone, whereas it was increased by glimepiride. Multiple stepwise regression analysis revealed that the increase in high-density lipoprotein cholesterol level was independently associated with the attenuation of plaque inflammation. CONCLUSIONS Our present study suggests that pioglitazone could attenuate atherosclerotic plaque inflammation in patients with impaired glucose tolerance or in diabetic patients independent of glucose lowering effect. Pioglitazone may be a promising strategy for the treatment of atherosclerotic plaque inflammation in impaired glucose tolerance or diabetic patients. (Detection of Plaque Inflammation and Visualization of Anti-Inflammatory Effects of Pioglitazone on Plaque Inflammation in Subjects With Impaired Glucose Tolerance and Type 2 Diabetes Mellitus by FDG-PET/CT; NCT00722631).


Journal of the American College of Cardiology | 2012

Cardiac-specific deletion of SOCS-3 prevents development of left ventricular remodeling after acute myocardial infarction.

Toyoharu Oba; Hideo Yasukawa; Masahiko Hoshijima; Ken-ichiro Sasaki; Nobuyoshi Futamata; Daisuke Fukui; Kazutoshi Mawatari; Takanobu Nagata; Sachiko Kyogoku; Hideki Ohshima; Tomoko Minami; Kei-ichiro Nakamura; Dongchon Kang; Toshitaka Yajima; Kirk U. Knowlton; Tsutomu Imaizumi

OBJECTIVES The study investigated the role of myocardial suppressor of cytokine signaling-3 (SOCS3), an intrinsic negative feedback regulator of the janus kinase and signal transducer and activator of transcription (JAK-STAT) signaling pathway, in the development of left ventricular (LV) remodeling after acute myocardial infarction (AMI). BACKGROUND LV remodeling after AMI results in poor cardiac performance leading to heart failure. Although it has been shown that JAK-STAT-activating cytokines prevent LV remodeling after AMI in animals, little is known about the role of SOCS3 in this process. METHODS Cardiac-specific SOCS3 knockout mice (SOCS3-CKO) were generated and subjected to AMI induced by permanent ligation of the left anterior descending coronary artery. RESULTS Although the initial infarct size after coronary occlusion measured by triphenyltetrazolium chloride staining was comparable between SOCS3-CKO and control mice, the infarct size 14 days after AMI was remarkably inhibited in SOCS3-CKO, indicating that progression of LV remodeling after AMI was prevented in SOCS3-CKO hearts. Prompt and marked up-regulations of multiple JAK-STAT-activating cytokines including leukemia inhibitory factor and granulocyte colony-stimulating factor (G-CSF) were observed within the heart following AMI. Cardiac-specific SOCS3 deletion enhanced multiple cardioprotective signaling pathways including STAT3, AKT, and extracellular signal-regulated kinase (ERK)-1/2, while inhibiting myocardial apoptosis and fibrosis as well as augmenting antioxidant expression. CONCLUSIONS Enhanced activation of cardioprotective signaling pathways by inhibiting myocardial SOCS3 expression prevented LV remodeling after AMI. Our data suggest that myocardial SOCS3 may be a key molecule in the development of LV remodeling after AMI.


Circulation | 2015

Renal Nerve-Mediated Erythropoietin Release Confers Cardioprotection During Remote Ischemic Preconditioning

Toyoharu Oba; Hideo Yasukawa; Takanobu Nagata; Sachiko Kyogoku; Tomoko Minami; Michihide Nishihara; Hideki Ohshima; Kazutoshi Mawatari; Shoichiro Nohara; Jinya Takahashi; Yusuke Sugi; Sachiyo Igata; Yoshiko Iwamoto; Hisashi Kai; Hidehiro Matsuoka; Makoto Takano; Hiroki Aoki; Yoshihiro Fukumoto; Tsutomu Imaizumi

BACKGROUND Remote ischemic preconditioning (RIPC) induced by transient limb ischemia is a powerful innate mechanism of cardioprotection against ischemia. Several described mechanisms explain how RIPC may act through neural pathways or humoral factors; however, the mechanistic pathway linking the remote organ to the heart has not yet been fully elucidated. This study aimed to investigate the mechanisms underlying the RIPC-induced production of Janus kinase (JAK)-signal transducer and activator of the transcription (STAT)-activating cytokines and cardioprotection by using mouse and human models of RIPC. METHODS AND RESULTS Screened circulating cardioprotective JAK-STAT-activating cytokines in mice unexpectedly revealed increased serum erythropoietin (EPO) levels after RIP induced by transient ischemia. In mice, RIPC rapidly upregulated EPO mRNA and its main transcriptional factor, hypoxia-inducible factor-1α (HIF1α), in the kidney. Laser Doppler blood flowmetry revealed a prompt reduction of renal blood flow (RBF) after RIPC. RIPC activated cardioprotective signaling pathways and the anti-apoptotic Bcl-xL pathway in the heart, and reduced infarct size. In mice, these effects were abolished by administration of an EPO-neutralizing antibody. Renal nerve denervation also abolished RIPC-induced RBF reduction, EPO production, and cardioprotection. In humans, transient limb ischemia of the upper arm reduced RBF and increased serum EPO levels. CONCLUSIONS Based on the present data, we propose a novel RIPC mechanism in which inhibition of infarct size by RIPC is produced through the renal nerve-mediated reduction of RBF associated with activation of the HIF1α-EPO pathway.


Circulation | 2011

Longest Survivor of Pulmonary Atresia With Ventricular Septal Defect Well-Developed Major Aortopulmonary Collateral Arteries Demonstrated by Multidetector Computed Tomography

Daisuke Fukui; Hisashi Kai; Tomohiro Takeuchi; Takeki Gondo; Toyoharu Oba; Kazutoshi Mawatari; Tatsuo Tonai; Yu Matsuo; Shin-ichiro Ueda; Hiroshi Niiyama; Takafumi Ueno; Tsutomu Imaizumi

A 59-year-old woman was admitted because of cyanosis and dyspnea on exertion and at rest. In her childhood, she was suspected of having ventricular septal defect (VSD), but she refused to undergo cardiac catheterization and operation. Dyspnea on exertion gradually developed after adolescence. On admission, chest roentgenography demonstrated enlarged cardiac silhouette with elevated cardiac apex, a right aortic arch, and enlargement of the main pulmonary arteries and their major branches with increased pulmonary arterial vascularity (Figure 1). Echocardiography revealed a large VSD which lay beneath the dilated aorta that overrides the interventricular septum, hypertrophied right ventricle, and the blind outflow tract of the right ventricle …


PLOS ONE | 2015

Cardiac-Specific SOCS3 Deletion Prevents In Vivo Myocardial Ischemia Reperfusion Injury through Sustained Activation of Cardioprotective Signaling Molecules.

Takanobu Nagata; Hideo Yasukawa; Sachiko Kyogoku; Toyoharu Oba; Jinya Takahashi; Shoichiro Nohara; Tomoko Minami; Kazutoshi Mawatari; Yusuke Sugi; Koutatsu Shimozono; Sylvain Pradervand; Masahiko Hoshijima; Hiroki Aoki; Yoshihiro Fukumoto; Tsutomu Imaizumi

Myocardial ischemia reperfusion injury (IRI) adversely affects cardiac performance and the prognosis of patients with acute myocardial infarction. Although myocardial signal transducer and activator of transcription (STAT) 3 is potently cardioprotective during IRI, the inhibitory mechanism responsible for its activation is largely unknown. The present study aimed to investigate the role of the myocardial suppressor of cytokine signaling (SOCS)-3, an intrinsic negative feedback regulator of the Janus kinase (JAK)-STAT signaling pathway, in the development of myocardial IRI. Myocardial IRI was induced in mice by ligating the left anterior descending coronary artery for 1 h, followed by different reperfusion times. One hour after reperfusion, the rapid expression of JAK-STAT–activating cytokines was observed. We precisely evaluated the phosphorylation of cardioprotective signaling molecules and the expression of SOCS3 during IRI and then induced myocardial IRI in wild-type and cardiac-specific SOCS3 knockout mice (SOCS3-CKO). The activation of STAT3, AKT, and ERK1/2 rapidly peaked and promptly decreased during IRI. This decrease correlated with the induction of SOCS3 expression up to 24 h after IRI in wild-type mice. The infarct size 24 h after reperfusion was significantly reduced in SOCS3-CKO compared with wild-type mice. In SOCS3-CKO mice, STAT3, AKT, and ERK1/2 phosphorylation was sustained, myocardial apoptosis was prevented, and the expression of anti-apoptotic Bcl-2 family member myeloid cell leukemia-1 (Mcl-1) was augmented. Cardiac-specific SOCS3 deletion led to the sustained activation of cardioprotective signaling molecules including and prevented myocardial apoptosis and injury during IRI. Our findings suggest that SOCS3 may represent a key factor that exacerbates the development of myocardial IRI.


International Journal of Cardiology | 2013

Screening for Fabry disease in patients with left ventricular hypertrophy

Kazutoshi Mawatari; Hideo Yasukawa; Toyoharu Oba; Takanobu Nagata; Tadayasu Togawa; Takahiro Tsukimura; Sachiko Kyogoku; Hideki Ohshima; Tomoko Minami; Yusuke Sugi; Hitoshi Sakuraba; Tsutomu Imaizumi

Kazutoshi Mawatari , Hideo Yasukawa ⁎, Toyoharu Oba , Takanobu Nagata , Tadayasu Togawa , Takahiro Tsukimura , Sachiko Kyogoku , Hideki Ohshima , Tomoko Minami , Yusuke Sugi , Hitoshi Sakuraba , Tsutomu Imaizumi a,b a Division of Cardiovascular Medicine, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan b Cardiovascular Research Institute, Kurume University, 67 Asahi-machi, Kurume 830-0011, Japan c Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan d Department of Clinical Genetics, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo 204-8588, Japan


Journal of Cardiac Failure | 2009

Cardiac-Specific Deletion of SOCS3 Prevented Myocardial Apoptosis After Acute Myocardial Infarction Through Inhibiting Mitochondrial Damage

Toyoharu Oba; Hideo Yasukawa; Ken-Ichirou Sasaki; Nobuyoshi Futamata; Kazutoshi Mawatari; Sachiko Kyougoku; Takanobu Nagata; Masahiko Hoshijima; Kirk U. Knowlton; Tsutomu Imaizumi


Circulation | 2016

Abstract 17237: Synergistic Reduction of Myocardial Ischemia Reperfusion Injury by Ischemic Preconditioning and Myocardial SOCS3 Deficiency in Mice

Shoichiro Nohara; Takanobu Nagata; Hideo Yasukawa; Yusuke Sugi; Kazutoshi Mawatari; Daisuke Fukui; Sachiko Kyogoku; Hideki Ohshima; Tomoko Minami; Jinya Takahashi; Koutatsu Shimozono; Tatsuhiro Shibata; Yoshihiro Fukumoto


Journal of Cardiac Failure | 2015

Change of Transcriptional Profile in Response to Cardiac-Specific Deletion of SOCS3 after Myocardial Ischemia Reperfusion Injury

Takanobu Nagata; Hideo Yasukawa; Jinya Takahashi; Koutatsu Shimozono; Shoichiro Nohara; Tomoko Minami; Toyoharu Oba; Kazutoshi Mawatari; Yusuke Sugi; Yoshihiro Fukumoto


Journal of Cardiac Failure | 2015

Cardiac-Specific SOCS3 Deficiency Leads to Left Ventricular Dysfunction with Advanced Age through Impaired Autophagy

Shoichiro Nohara; Hideo Yasukawa; Toyoharu Oba; Takanobu Nagata; Jinya Takahashi; Koutatsu Shimozono; Tomoko Minami; Kazutoshi Mawatari; Yusuke Sugi; Yoshihiro Fukumoto

Collaboration


Dive into the Kazutoshi Mawatari'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