Network


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

Hotspot


Dive into the research topics where Yukihiro Takemura is active.

Publication


Featured researches published by Yukihiro Takemura.


Endocrine | 2009

Klotho suppresses TNF-α-induced expression of adhesion molecules in the endothelium and attenuates NF-κB activation

Yoshihiro Maekawa; Kazuhiko Ishikawa; Osamu Yasuda; Ryosuke Oguro; Hiroko Hanasaki; Iwao Kida; Yukihiro Takemura; Mitsuru Ohishi; Tomohiro Katsuya; Hiromi Rakugi

Klotho is a senescence suppressor protein that, when overexpressed, extends the lifespan of mice. Klotho-disrupted mice exhibit atherosclerosis and endothelial dysfunction, which led us to investigate the effect of the Klotho protein on vascular inflammation, particularly adhesion molecule expression. In this study, human umbilical vein endothelial cells (HUVECs) were preincubated with Klotho protein and then exposed to tumor necrosis factor-α (TNF-α) or vehicle. Reverse transcription-PCR and Western blot analyses revealed that Klotho suppressed TNF-α-induced expression of intracellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1). NF-κB activation, IκB phosphorylation induced by TNF-α were also attenuated by Klotho protein administration. The inhibition of eNOS phosphorylation by TNF-α was reversed by Klotho. Furthermore, Klotho inhibited TNF-α-induced monocyte adhesion to HUVECs and suppressed adhesion molecule expression in an organ culture of the rat aorta. These results suggest that Klotho suppresses TNF-α-induced expression of adhesion molecules and NF-κB activation. Klotho may have a role in the modulation of endothelial inflammation.


Diabetes | 2006

Apoptosis Signal-Regulating Kinase 1 Mediates Cellular Senescence Induced by High Glucose in Endothelial Cells

Toyohiko Yokoi; Keisuke Fukuo; Osamu Yasuda; Mizuo Hotta; Jun-ichi Miyazaki; Yukihiro Takemura; Hidenobu Kawamoto; Hidenori Ichijo; Toshio Ogihara

Vascular ageing is accelerated in patients with diabetes. However, the underlying mechanism remains unclear. Here, we show that high glucose induces activation of apoptosis signal-regulating kinase 1 (ASK1), an apoptosis-inducing signal that mediates endothelial cell senescence induced by hyperglycemia. High glucose induced a time-dependent increase in the levels of ASK1 expression and its activity in human umbilical vein endothelial cells (HUVECs). Incubation of endothelial cells with high glucose increased the proportion of cells expressing senescence-associated β-galactosidase (SA-β-gal) activity. However, transfection with an adenoviral construct including a dominant negative form of ASK1 gene significantly inhibited SA-β-gal activity induced by high glucose. In addition, infection with an adenoviral construct expressing the constitutively active ASK1 gene directly induced an increase in the levels of SA-β-gal activity. Activation of the ASK1 signal also enhanced plasminogen activator inhibitor-1 (PAI-1) expression in HUVECs. Induction of senescent endothelial cells in aortas and elevation of plasma PAI-1 levels were observed in streptozotocin (STZ) diabetic mice, whereas these changes induced by STZ were attenuated in ASK1-knockout mice. Our results suggest that hyperglycemia accelerates endothelial cell senescence and upregulation of PAI-1 expression through activation of the ASK1 signal. Thus, ASK1 may be a new therapeutic target to prevent vascular ageing and thrombosis in diabetic patients.


Hypertension | 2004

Homocysteine Enhances Endothelial Apoptosis via Upregulation of Fas-Mediated Pathways

Toshimitsu Suhara; Keisuke Fukuo; Osamu Yasuda; Maki Tsubakimoto; Yukihiro Takemura; Hidenobu Kawamoto; Toyohiko Yokoi; Masaki Mogi; Taeko Kaimoto; Toshio Ogihara

Hyperhomocysteinemia is an independent risk factor for the development of atherosclerosis. However, the underlying mechanism of endothelial cell injury in hyperhomocysteinemia has not been elucidated. In this study, we examined the effect of homocysteine (Hcy) on Fas-mediated apoptosis in endothelial cells. Hcy-induced upregulation of Fas in endothelial cells (ECs) in a dose-dependent manner. At the same time, Hcy increased intracellular peroxide in ECs. Hcy-induced Fas expression was inhibited by the treatment with catalase. Hcy increased NF-κinding activity, and adenovirus-mediated transfection of a I κ-B mutant (Iκ-B mt) gene inhibited Hcy-induced Fas expression. ECs were sensitive to Fas-mediated apoptosis when exposed to Hcy. Under these condition, Iκ-B mt protected ECs from Fas-mediated apoptosis. In addition, Hcy inhibited expression of the caspase-8 inhibitor FLICE-inhibitory protein (FLIP). Adenovirus-mediated transfection of constitutively active Akt gene abolished the Hcy-mediated downregulation of FLIP. These data suggest that upregulation of Fas expression and downregulation of FLIP is a mechanism through which Hcy induces EC apoptosis.


Diabetes | 2013

Loss of ACE2 Exaggerates High-Calorie Diet–Induced Insulin Resistance by Reduction of GLUT4 in Mice

Masao Takeda; Koichi Yamamoto; Yukihiro Takemura; Hikari Takeshita; Kazuhiro Hongyo; Tatsuo Kawai; Hiroko Hanasaki-Yamamoto; Ryosuke Oguro; Yoichi Takami; Yuji Tatara; Yasushi Takeya; Ken Sugimoto; Kei Kamide; Mitsuru Ohishi; Hiromi Rakugi

ACE type 2 (ACE2) functions as a negative regulator of the renin-angiotensin system by cleaving angiotensin II (AII) into angiotensin 1–7 (A1–7). This study assessed the role of endogenous ACE2 in maintaining insulin sensitivity. Twelve-week-old male ACE2 knockout (ACE2KO) mice had normal insulin sensitivities when fed a standard diet. AII infusion or a high-fat, high-sucrose (HFHS) diet impaired glucose tolerance and insulin sensitivity more severely in ACE2KO mice than in their wild-type (WT) littermates. The strain difference in glucose tolerance was not eliminated by an AII receptor type 1 (AT1) blocker but was eradicated by A1–7 or an AT1 blocker combined with the A1–7 inhibitor (A779). The expression of GLUT4 and a transcriptional factor, myocyte enhancer factor (MEF) 2A, was dramatically reduced in the skeletal muscles of the standard diet–fed ACE2KO mice. The expression of GLUT4 and MEF2A was increased by A1–7 in ACE2KO mice and decreased by A779 in WT mice. A1–7 enhanced upregulation of MEF2A and GLUT4 during differentiation of myoblast cells. In conclusion, ACE2 protects against high-calorie diet–induced insulin resistance in mice. This mechanism may involve the transcriptional regulation of GLUT4 via an A1–7–dependent pathway.


Cellular and Molecular Life Sciences | 2008

Aspirin: recent developments

Osamu Yasuda; Yukihiro Takemura; Hidenobu Kawamoto; Hiromi Rakugi

Abstract.Aspirin exerts anti-thrombotic action by acetylating and inactivating cyclooxygenase-1, preventing the production of thromboxane A2 in platelets. Through this inhibition of platelet function, aspirin is considered as a preventative of ischemic diseases such as coronary and cerebral infarction. However, many studies have revealed that aspirin has other beneficial actions in addition to its anti-platelet activity. For example, aspirin may confer some benefit against colorectal cancer. Here, we discuss the involvement of inflammation in atherosclerosis and how aspirin exerts its beneficial actions in atherosclerotic diseases and cancer.


Hypertension | 2003

Eicosapentaenoic Acid Protects Endothelial Cells Against Anoikis Through Restoration of cFLIP

Takashi Suzuki; Keisuke Fukuo; Toshimitsu Suhara; Osamu Yasuda; Naoyuki Sato; Yukihiro Takemura; Maki Tsubakimoto; Toshio Ogihara

Abstract—Dietary supplementation with eicosapentaenoic acid (EPA) improves the prognosis of chronic inflammatory diseases, including atherosclerosis. The mechanism underlying these beneficial effects, however, remains to be elucidated. Here we show that EPA protects endothelial cells from anoikis through upregulation of the cellular FLICE (Fas-associating protein with death domain-like interleukin-1–converting enzyme)-inhibitory protein (cFLIP), an endogenous inhibitor of caspase-8. EPA-induced upregulation of cFLIP expression was partially suppressed by the phosphatidylinositol-3-kinase inhibitor wortmannin. Conversely, treatment with insulinlike growth factor-1 (IGF-1), an activator of phosphatidylinositol-3-kinase/Akt signaling, or infection with an adenoviral construct expressing the constitutively active Akt gene induced upregulation of cFLIP expression. In addition, pretreatment of endothelial cells with either EPA or IGF-1 protected them from anoikis, suggesting that EPA-induced protection against anoikis is partially mediated through activation of Akt. On the other hand, when endothelial cells were already detached, treatment of these cells with EPA but not with IGF-1 protected them against anoikis. Importantly, EPA restored cFLIP expression without activating Akt signaling in detached endothelial cells, whereas IGF-1 had no effect. Additionally, exogenously restored expression of cFLIP by the tetracycline-regulated adenovirus system protected endothelial cells against anoikis. Furthermore, EPA was protective against the loss of endothelium in an organ culture of rat aortas. These findings suggest that EPA protects against endothelial cell anoikis through restoration of cFLIP expression, which might contribute to the mechanism underlying the beneficial effects of EPA in patients with hypertension.


Hypertension | 2004

Fas Signaling Induces Akt Activation and Upregulation of Endothelial Nitric Oxide Synthase Expression

Yukihiro Takemura; Keisuke Fukuo; Osamu Yasuda; Takahito Inoue; Norio Inomata; Toyohiko Yokoi; Hidenobu Kawamoto; Toshimitsu Suhara; Toshio Ogihara

Abstract—A growing body of evidence has shown that Fas, a death receptor, mediates apoptosis-unrelated biological effects. Here, we report that Fas engagement with Fas ligand induced activation of Akt and upregulation of endothelial nitric oxide synthase expression without induction of apoptosis. In the presence of the phosphatidylinositol 3-kinase inhibitor wortmannin, Fas ligand, however, induced apoptosis instead of upregulation of endothelial nitric oxide synthase expression. In vivo, systolic blood pressure was slightly higher in mutant mice with decreased cell surface Fas expression (lpr mice) compared with wild-type mice. In addition, chronic inhibition of nitric oxide synthesis by NG-nitro-l-arginine induced a progressive increase in the levels of blood pressure in wild-type mice, whereas no further increase in the levels of blood pressure was observed in lpr mice. Furthermore, acetylcholine caused a lesser endothelium-dependent relaxation of the strips from lpr mice compared with wild-type mice, although the vasoconstrictor potency of phenylephrine was not different between the two groups. These findings indicate that Fas signaling may have a role in the regulation of endothelial function and blood pressure through modulating endothelial nitric oxide synthase expression in the Akt signal-dependent manner.


Hypertension | 2010

Nifedipine Inhibits Vascular Smooth Muscle Cell Dedifferentiation via Downregulation of Akt Signaling

Taeko Kaimoto; Osamu Yasuda; Mitsuru Ohishi; Masaki Mogi; Yukihiro Takemura; Toshimitsu Suhara; Toshio Ogihara; Keisuke Fukuo; Hiromi Rakugi

Calcium is an essential signaling molecule that controls vascular smooth muscle cell (VSMC) contraction, proliferation, and differentiation. Here, we show that the calcium antagonist nifedipine inhibits VSMC dedifferentiation in vitro and in vivo. Differentiated VSMCs cultured on laminin-coated dishes were transferred to laminin-free dishes to induce dedifferentiation. Induction of dedifferentiation resulted in the upregulation of nonmuscle myosin heavy chain expression, a marker of dedifferentiation, and the downregulation of smooth muscle myosin heavy chain expression, a marker of differentiation. Nifedipine significantly inhibited both the induction of these phenotypic changes and upregulation of Akt signaling in these cells. Administration of nifedipine at a low concentration that did not affect blood pressure could inhibit the increase in nonmuscle myosin heavy chain expression and decrease in smooth muscle myosin heavy chain expression in a rat balloon-injury model. Furthermore, nifedipine suppressed neointimal hyperplasia and upregulation of Akt signaling. However, phospho-Akt expression was not suppressed in the regenerating arterial endothelium of the nifedipine-treated rats. The inhibitory effect of the downregulation of Akt signaling by dominant-negative Akt on the induction of VSMC dedifferentiation in the intima was identical to that of nifedipine. In contrast, upregulation of Akt signaling by transfection of the cells with a constitutively active Akt reversed the nifedipine-induced inhibition of VSMC dedifferentiation. In conclusion, nifedipine inhibits VSMC dedifferentiation by suppressing Akt signaling, thereby preventing neointimal thickening.


Hypertension Research | 2009

Fas promoter region gene polymorphism is associated with an increased risk for myocardial infarction

Hiroko Hanasaki; Yukihiro Takemura; Keisuke Fukuo; Mitsuru Ohishi; Miyuki Onishi; Osamu Yasuda; Tomohiro Katsuya; Nobuhisa Awata; Norihiro Kato; Toshio Ogihara; Hiromi Rakugi

A growing body of evidence has shown that Fas-mediated apoptosis is involved in atherosclerosis progression. Recent studies have revealed that a single nucleotide polymorphism (SNP) in the Fas promoter region (−670G/A) influences Fas expression. Here, we investigated whether −670G/A SNP influences the incidence of myocardial infarction (MI) by examining a comparison between MI patients (n=154) and control subjects (n=462) in a Japanese population. The allele frequency in each group was A 53.6%/G 46.4% in the MI patients, and A 43.9%/G 56.1% in the non-MI subjects (χ2=8.6; P=0.003). The odds ratio was 2.62 (95% CI: 1.43–4.88). As subjects with the −670AA genotype had a signal transducer and activator of transcription 1 (STAT1)-binding site in the Fas promoter region, STAT-1 activation by interferon-γ may upregulate Fas expression in human vascular smooth muscle cells (VSMCs) of −670AA genotype subjects as described earlier. The Fas upregulation induces excess apoptosis to VSMCs, which leads to unstable plaque formation in atherosclerotic lesions and then potentially to plaque rupture, which can cause MI. Further investigation of hypertensive subjects revealed that the −670AA genotype does not induce hypertension occurrence, supporting that this difference of MI occurrence between the −670AA genotype and the −670GG genotype may be because of plaque rupture followed by excess apoptosis of VSMCs in the atherosclerotic lesion. We conclude that the Fas promoter gene, SNP (−670G/A), may be a risk factor of MI occurrence.


Biochemical and Biophysical Research Communications | 2008

Akt activation prevents Apop-1-induced death of cells

Xin Sun; Osamu Yasuda; Yukihiro Takemura; Hidenobu Kawamoto; Masayoshi Higuchi; Yoshichika Baba; Tomohiro Katsuya; Keisuke Fukuo; Toshio Ogihara; Hiromi Rakugi

Apop-1 is a novel protein identified in cultured atherosclerotic smooth muscle cells of ApoE-deficient mice, and the expression of the Apop-1 protein induces the death of cultured cells. Insulin-like growth factor-1 (IGF-1) is a well-characterized survival factor for VSMC; however, the interaction between Apop-1 and survival factor IGF-1 in the mediation of cell death is poorly understood. In this report, we show that the IGF-1 signaling cascade protects VSMC against Apop-1-induced death. Furthermore, our data indicate that the inhibition of Apop-1-induced death by IGF-1 is mediated by the activation of the PI3K/Akt signaling pathway.

Collaboration


Dive into the Yukihiro Takemura'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