Network


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

Hotspot


Dive into the research topics where Tetsuya Hisada is active.

Publication


Featured researches published by Tetsuya Hisada.


Biochemical Pharmacology | 1999

Inhibitory effect of tea flavonoids on the ability of cells to oxidize low density lipoprotein

Hiroshi Yoshida; Toshitsugu Ishikawa; Hiroshi Hosoai; Michio Suzukawa; Makoto Ayaori; Tetsuya Hisada; Shojiro Sawada; Atsushi Yonemura; Kenji Higashi; Toshimitsu Ito; Kei Nakajima; Takeshi Yamashita; Koji Tomiyasu; Masato Nishiwaki; Fumitaka Ohsuzu; Haruo Nakamura

Dietary flavonoid intake has been reported to be inversely related to mortality from coronary heart disease, and the anti-atherosclerotic effect of flavonoids is considered to be due probably to their antioxidant properties. Oxidation of low density lipoprotein (LDL) has been reported to be induced by the constituent cells of the arterial wall. Accordingly, we examined the effect of pretreatment with tea flavonoids, such as theaflavin digallate, on the ability of cells to oxidize LDL. Theaflavin digallate pretreatment of macrophages or endothelial cells reduced cell-mediated LDL oxidation in a concentration- (0-400 microM) and time- (0-4 hr) dependent manner. This inhibitory effect of flavonoids on cell-mediated LDL oxidation was in the order of theaflavin digallate > theaflavin > or = epigallocatechin gallate > epigallocatechin > gallic acid. Further, we investigated the mechanisms by which flavonoids inhibited cell-mediated LDL oxidation using macrophages and theaflavin digallate. Theaflavin digallate pretreatment decreased superoxide production of macrophages and chelated iron ions significantly. These results suggest that tea flavonoids attenuate the ability of the cell to oxidize LDL, probably by reducing superoxide production in cells and chelating iron ions.


Journal of Biological Chemistry | 2003

Role of the Glucocorticoid Receptor for Regulation of Hypoxia-dependent Gene Expression

Tsunenori Kodama; Noriaki Shimizu; Noritada Yoshikawa; Yuichi Makino; Rika Ouchida; Kensaku Okamoto; Tetsuya Hisada; Hiroshi Nakamura; Chikao Morimoto; Hirotoshi Tanaka

Glucocorticoids are secreted from the adrenal glands and act as a peripheral effector of the hypothalamic-pituitary-adrenal axis, playing an essential role in stress response and homeostatic regulation. In target cells, however, it remains unknown how glucocorticoids finetune the cellular pathways mediating tissue and systemic adaptation. Recently, considerable evidence indicates that adaptation to hypoxic environments is influenced by glucocorticoids and there is cross-talk between hypoxia-dependent signals and glucocorticoid-mediated regulation of gene expression. We therefore investigated the interaction between these important stress-responsive pathways, focusing on the glucocorticoid receptor (GR) and hypoxia-inducible transcription factor HIF-1. Here we show that, under hypoxic conditions, HIF-1-dependent gene expression is further up-regulated by glucocorticoids via the GR. This up-regulation cannot be substituted by the other steroid receptors and is suggested to result from the interaction between the GR and the transactivation domain of HIF-1α. Moreover, our results also indicate that the ligand binding domain of the GR is essential for this interaction, and the critical requirement for GR agonists suggests the importance of the ligand-mediated conformational change of the GR. Because these proteins are shown to colocalize in the distinct compartments of the nucleus, we suggest that these stress-responsive transcription factors have intimate communication in close proximity to each other, thereby enabling the fine-tuning of cellular responses for adaptation.


Circulation Research | 2010

Coffee Consumption Enhances High-Density Lipoprotein-Mediated Cholesterol Efflux in Macrophages

Harumi Uto-Kondo; Makoto Ayaori; Masatsune Ogura; Kazuhiro Nakaya; Mai Ito; Atsushi Suzuki; Shunichi Takiguchi; Emi Yakushiji; Yoshio Terao; Hideki Ozasa; Tetsuya Hisada; Makoto Sasaki; Fumitaka Ohsuzu; Katsunori Ikewaki

Rationale: Association of habitual coffee consumption with coronary heart disease morbidity and mortality has not been established. We hypothesized that coffee may enhance reverse cholesterol transport (RCT) as the antiatherogenic properties of high-density lipoprotein (HDL). Objective: This study was to investigate whether the phenolic acids of coffee and coffee regulates RCT from macrophages in vitro, ex vivo and in vivo. Methods and Results: Caffeic acid and ferulic acid, the major phenolic acids of coffee, enhanced cholesterol efflux from THP-1 macrophages mediated by HDL, but not apoA-I. Furthermore, these phenolic acids increased both the mRNA and protein levels of ATP-binding cassette transporter (ABC)G1 and scavenger receptor class B type I (SR-BI), but not ABCA1. Eight healthy volunteers were recruited for the ex vivo study, and blood samples were taken before and 30 minutes after consumption of coffee or water in a crossover study. The mRNA as well as protein levels of ABCG1, SR-BI, and cholesterol efflux by HDL were increased in the macrophages differentiated under autologous sera obtained after coffee consumption compared to baseline sera. Finally, effects of coffee and phenolic acid on in vivo RCT were assessed by intraperitoneally injecting [3H]cholesterol-labeled acetyl low-density lipoprotein–loaded RAW264.7 cells into mice, then monitoring appearance of 3H tracer in plasma, liver, and feces. Supporting in vitro and ex vivo data, ferulic acid was found to significantly increase the levels of 3H tracer in feces. Conclusions: Coffee intake might have an antiatherogenic property by increasing ABCG1 and SR-BI expression and enhancing HDL-mediated cholesterol efflux from the macrophages via its plasma phenolic acids.


Genes to Cells | 2003

Suppression of NF-kappaB-dependent gene expression by a hexamethylene bisacetamide-inducible protein HEXIM1 in human vascular smooth muscle cells.

Rika Ouchida; Masatoshi Kusuhara; Noriaki Shimizu; Tetsuya Hisada; Yuichi Makino; Chikao Morimoto; Hiroshi Handa; Fumitaka Ohsuzu; Hirotoshi Tanaka

Background: Neointima formation is a characteristic feature of atherosclerosis and post‐angioplasty restenosis, in which various soluble factors and mechanical injury stimulate signalling pathways in vascular smooth muscle cells (VSMC), promoting their migration and proliferation, and the eventual formation of the neointima. The transcription factor NF‐κB has been shown to play a pivotal role in this process. Hexamethylene bisacetamide, an inhibitor of VSMC proliferation, induces the mRNA expression of HEXIM1 (hexamethylene bisacetamide‐inducible protein 1). However, the protein expression and function of HEXIM1 remain unknown.


Pflügers Archiv: European Journal of Physiology | 1990

Properties of membrane currents in isolated smooth muscle cells from guinea-pig trachea

Tetsuya Hisada; Yoshihisa Kurachi; Tsuneaki Sugimoto

Tracheal smooth muscle cells were enzymatically isolated from guinea-pig trachea. These cells contracted in response to acetylcholine (0.01–10 μM) in a concentration-dependent fashion. Under current-clamp conditions with 140 mM K+ in the pipette solution, the membrane potential oscillated spontaneously at around −30 mV. Under voltage-clamp conditions, there appeared spontaneous but steady oscillations of outward current (Io). On depolarization from a holding potential at −40 mV, three components of outward current were elicited: transient outward current (IT), steady-state outward current (Is) and Io. These three components of outward current reversed around the K+ equilibrium potential and were abolished by Cs+ in the pipette, indicating that K+ was the major charge carrier of these outward currents. All these three components were completely suppressed by extracellular tetraethylammonium (10 mM). Both IT and Io were depressed by quinidine (1 mM), 4-aminopyridine (10 mM) and nifedipine (100 nM), but Is was not affected. IT and Io were suppressed by a Ca2+-free perfusate with less than 1 nM Ca2+ in the pipette, while with 10 nM Ca2+ in the pipette, only Io was suppressed. In both conditions, Is was not affected by the Ca2+-free perfusate. Therefore, it is suggested that Io, IT and Is are separate types of K+ current. With Cs+ in the pipette, K+ currents were almost completely suppressed and a transient inward current was observed during depolarizing pulses. The inward current was not affected by tetrodotoxin and increased when the concentration of extracellular Ca2+ was raised, indicating that the current is a Ca2+ channel current. Even with a holding potential of −80 mV, the low-threshold inward current could not be observed. The high-threshold Ca2+ current was abolished by nifedipine (100 nM) and was enhanced by Bay K 8644 (100 nM). The order of permeation of divalent cations through the Ca2+ channel was Ba2+ >Sr2+ ≈ Ca2+. Cd2+ blocked the Ca2+ current more effectively than Ni2+. These results may indicate that the Ca2+ current of tracheal smooth muscle cells is mainly composed of the current through an L-type Ca2+ channel.


Atherosclerosis | 2011

Pioglitazone enhances cholesterol efflux from macrophages by increasing ABCA1/ABCG1 expressions via PPARγ/LXRα pathway: Findings from in vitro and ex vivo studies

Hideki Ozasa; Makoto Ayaori; Maki Iizuka; Yoshio Terao; Harumi Uto-Kondo; Emi Yakushiji; Shunichi Takiguchi; Kazuhiro Nakaya; Tetsuya Hisada; Yoshinari Uehara; Masatsune Ogura; Makoto Sasaki; Tomohiro Komatsu; Shunpei Horii; Seibu Mochizuki; Michihiro Yoshimura; Katsunori Ikewaki

OBJECTIVE Pioglitazone, a peroxisome proliferator-activated receptor γ (PPARγ) agonist, reportedly reduces cardiovascular events in diabetic patients. ATP cassette binding transporters (ABC) A1 and G1 are pivotal molecules for cholesterol efflux (ChE) from macrophages and high density-lipoprotein biogenesis, and the A1 transporter is regulated by a PPARγ-liver receptor X (LXR) pathway. Also, pioglitazone induces ABCG1 expression, though the exact mechanism remains unclear. We therefore investigated the effects of pioglitazone on ABCA1/G1 expression in vitro and ex vivo. METHODS The effects of pioglitazone on ChE and ABCA1/G1 expressions in macrophages were assessed. Then, mRNA was quantified in macrophages when PPARγ/LXR inhibition by siRNA or overexpression of oxysterol sulfotransferase was performed. ABCA1/G1 promoter activity with mutated LXR-responsive elements was also measured. As an ex vivo study, 15 type 2 diabetic patients were administered pioglitazone or placebo, and ChE assays and protein expressions were determined using macrophages cultured with the corresponding sera. RESULTS Pioglitazone increased LXRα/ABCA1/G1 expressions, which enhanced ChE from macrophages. Inhibition of PPARγ/LXR pathways revealed that LXR was primarily involved in pioglitazones transactivation of ABCA1 but only partially involved for ABCG1. Promoter assays showed that ABCG1 was regulated more by the promoter in intron 4 than that upstream of exon 1 but both promoters were responsive to LXR activation. Sera obtained after pioglitazone treatment promoted ChE and ABCA1/G1 expressions in macrophages. CONCLUSION Pioglitazone enhanced ChE from macrophages by increasing ABCA1/G1 in LXR-dependent and -independent manners. Our comparable in vitro and ex vivo results shed new light on pioglitazones novel anti-atherogenic property.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

Proteasomal Inhibition Promotes ATP-Binding Cassette Transporter A1 (ABCA1) and ABCG1 Expression and Cholesterol Efflux From Macrophages In Vitro and In Vivo

Masatsune Ogura; Makoto Ayaori; Yoshio Terao; Tetsuya Hisada; Maki Iizuka; Shunichi Takiguchi; Harumi Uto-Kondo; Emi Yakushiji; Kazuhiro Nakaya; Makoto Sasaki; Tomohiro Komatsu; Hideki Ozasa; Fumitaka Ohsuzu; Katsunori Ikewaki

Objective— ATP-binding cassette transporter A1 (ABCA1) and ABCG1 are key molecules in an initial step of reverse cholesterol transport (RCT), a major antiatherogenic property of high-density lipoprotein (HDL). The ubiquitin-proteasome system (UPS) mediates nonlysosomal pathways for protein degradation and is known to be involved in atherosclerosis. However, little is known about the effects of the UPS on these molecules and overall RCT. We therefore investigated whether UPS inhibition affects ABCA1/G1 expression in macrophages and RCT in vitro and in vivo. Methods and Results— Various proteasome inhibitors increased ABCA1/G1 expression in macrophages, translating into enhanced apolipoprotein A-I– and HDL-mediated cholesterol efflux from macrophages. ABCA1 and ABCG1 were found to undergo polyubiquitination in the macrophages and HEK293 cells overexpressing these proteins, and pulse-chase analysis revealed that proteasome inhibitors inhibited ABCA1/G1 protein degradation. In in vivo experiments, the proteasome inhibitor bortezomib increased ABCA1/G1 protein levels in mouse peritoneal macrophages, and RCT assays showed that it significantly increased the fecal (54% increase compared with saline) and plasma (23%) appearances of the tracer derived from intraperitoneally injected 3H-cholesterol-labeled macrophages. Conclusion— The present study provided evidence that the UPS is involved in ABCA1/G1 degradation, thereby affecting RCT in vivo. Therefore, specific inhibition of the UPS pathway might lead to a novel HDL therapy that enhances RCT.


Atherosclerosis | 2010

Cilostazol enhances macrophage reverse cholesterol transport in vitro and in vivo

Kazuhiro Nakaya; Makoto Ayaori; Harumi Uto-Kondo; Tetsuya Hisada; Masatsune Ogura; Emi Yakushiji; Shunichi Takiguchi; Yoshio Terao; Hideki Ozasa; Makoto Sasaki; Tomohiro Komatsu; Fumitaka Ohsuzu; Katsunori Ikewaki

OBJECTIVE Recent failure of an HDL-cholesterol raising strategy using a cholesteryl ester transfer protein inhibitor highlights the importance of the anti-atherogenic function rather than plasma concentration of HDL. Cilostazol, a selective inhibitor of phosphodiesterase 3, has been widely used in patients with atherosclerotic diseases and is known to increase HDL-cholesterol. However, it remains unclear whether cilostazol enhances anti-atherogenic properties by promoting reverse cholesterol transport (RCT), a major anti-atherogenic function of HDL. METHODS AND RESULTS We observed that treatment of THP-1 macrophages, human monocyte-derived macrophages, and RAW264.7 cells with cilostazol increased ABCA1 and ABCG1 expression in a concentration-dependent manner, translating into enhanced apoA-I- and HDL-mediated cholesterol efflux from the macrophages. However, other cyclic AMP (cAMP)-elevating agents did not increase ABCA1 gene expression in THP-1 macrophages. Cilostazol did not change intracellular cAMP levels in THP-1 macrophages and RAW264.7 cells, and a protein kinase A (PKA) inhibitor did not affect cilostazol-induced ABCA1 and ABCG1 expression. To further investigate RCT in vivo, (3)H-cholesterol-labeled and acetyl LDL-loaded RAW264.7 cells were intraperitoneally injected into mice and the appearance of the (3)H-tracer was monitored in plasma, liver, and feces. Supporting the in vitro data, cilostazol was found to significantly increase (3)H-tracer levels in both plasma and feces. CONCLUSIONS These findings indicate that cilostazol might provide anti-atherosclerotic effects by promoting RCT through increased ABCA1/G1 expression in macrophages.


Biochimica et Biophysica Acta | 2012

Retinoic acid receptor agonists regulate expression of ATP-binding cassette transporter G1 in macrophages.

Makoto Ayaori; Emi Yakushiji; Masatsune Ogura; Kazuhiro Nakaya; Tetsuya Hisada; Harumi Uto-Kondo; Shunichi Takiguchi; Yoshio Terao; Makoto Sasaki; Tomohiro Komatsu; Maki Iizuka; Makiko Yogo; Yoshinari Uehara; Hiroyuki Kagechika; Tsuyoshi Nakanishi; Katsunori Ikewaki

ABC transporter G1 (ABCG1) plays a pivotal role in HDL-mediated cholesterol efflux and atherogenesis. We investigated whether, and how, retinoic acid receptors (RARs) regulate ABCG1 expression in macrophages. All-trans retinoic acid (ATRA), an RAR ligand, increased ABCG1 protein levels and apoA-I/HDL-mediated cholesterol efflux from the macrophages. Both ATRA and other RAR agonists, TTNPB and Am580, increased major transcripts driven by promoter B upstream of exon 5, though minor transcripts driven by promoter A upstream of exon 1 were only increased by ATRA. The stimulatory effects of ATRA on ABCG1 expression were completely abolished in the presence of RAR/RXR antagonists but were only partially canceled in the presence of an LXR antagonist. Adenovirus with overexpressed oxysterol sulfotransferase abolished the LXR pathway, as previously reported, and ATRA-responsiveness in ABCA1/ABCG1 expressions were respectively attenuated by 38 and 22% compared to the control virus. Promoter assays revealed that ABCG1 levels were regulated more by promoter B than promoter A, and ATRA activated promoter B in a liver X receptor-responsive element (LXRE)-dependent manner. Further, LXRE-B in intron 7, but not LXRE-A in intron 5, enhanced ATRA responsiveness under overexpression of all RAR isoforms-RARα/β/γ. In contrast, the activation of promoter B by TTNPB depended on LXRE-B and RARα, but not on RARβ/γ. Finally, chromatin immunoprecipitation and gel-shift assays revealed a specific and direct repeat 4-dependent binding of RARα to LXRE-B. In conclusion, RAR ligands increase ABCA1/G1 expression and apoA-I/HDL-mediated cholesterol efflux from macrophages, and modulate ABCG1 promoter activity via LXRE-dependent mechanisms.


Cardiovascular Research | 2012

Statin Inhibits Hypoxia-Induced Endothelin-1 via accelerated degradation of HIF-1α in Vascular Smooth Muscle Cells

Tetsuya Hisada; Makoto Ayaori; Nobuhiro Ohrui; Hiroshi Nakashima; Kazuhiro Nakaya; Harumi Uto-Kondo; Emi Yakushiji; Shunichi Takiguchi; Yoshio Terao; Yoshinori Miyamoto; Takeshi Adachi; Haruo Nakamura; Fumitaka Ohsuzu; Katsunori Ikewaki; Yutaka Sakurai

AIMS Endothelin-1 (ET-1) contributes to the pathogenesis of cardiovascular diseases with multiple properties such as vasoconstriction. Human ET-1 gene expression is up-regulated by the transcription factor hypoxia-inducible factor-1 (HIF-1) through hypoxia response element (HRE). Although previous studies suggested that 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) alter HIF-1-related gene expression, it remained unclear whether statins modulate HIF-1-mediated ET-1 expression. Therefore, we investigated the effect of fluvastatin on hypoxia-induced human ET-1 expression in vascular smooth muscle cells (VSMC). METHODS AND RESULTS Hypoxia (1% O(2)), compared with the normoxic condition (21% O(2)), significantly induced the expression of preproET-1 mRNA, ET-1 protein, and ET-1 secretion in VSMC. Hypoxia induced a 2.3-fold increase in HRE-dependent ET-1 reporter gene activation. Under concentrations of 1 µmol/L or greater, fluvastatin attenuated the hypoxia-induced ET-1 gene expression through the accelerated ubiquitin/proteasome-dependent degradation of HIF-1α, thus consequently attenuating HIF-1α binding to the HRE of the ET-1 gene. These inhibitory effects of fluvastatin were cancelled by concomitant treatment with mevalonate, farnesyl pyrophosphate, or geranylgeranyl pyrophosphate, but not squalene. CONCLUSION The present study suggests that fluvastatin attenuates HIF-1-dependent ET-1 gene expression in conjunction with the stimulation of HIF-1α ubiquitin/proteasome-dependent degradation via isoprenoid-dependent mechanisms.

Collaboration


Dive into the Tetsuya Hisada's collaboration.

Top Co-Authors

Avatar

Makoto Ayaori

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Kazuhiro Nakaya

Asahikawa Medical University

View shared research outputs
Top Co-Authors

Avatar

Emi Yakushiji

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Katsunori Ikewaki

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Masatsune Ogura

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Shunichi Takiguchi

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Fumitaka Ohsuzu

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Harumi Uto-Kondo

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Haruo Nakamura

National Defense Medical College

View shared research outputs
Top Co-Authors

Avatar

Makoto Sasaki

National Defense Medical College

View shared research outputs
Researchain Logo
Decentralizing Knowledge