Network


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

Hotspot


Dive into the research topics where Soichiro Fujiyama is active.

Publication


Featured researches published by Soichiro Fujiyama.


Journal of Clinical Investigation | 1999

Angiotensin II type 2 receptor overexpression activates the vascular kinin system and causes vasodilation

Yoshiaki Tsutsumi; Hiroaki Matsubara; Hiroya Masaki; Hiroki Kurihara; Satoshi Murasawa; Shinji Takai; Mizuo Miyazaki; Yoshihisa Nozawa; Ryoji Ozono; Keigo Nakagawa; Takeshi Miwa; Noritaka Kawada; Yasukiyo Mori; Yasunobu Shibasaki; Yohko Tanaka; Soichiro Fujiyama; Yohko Koyama; Atsuko Fujiyama; Hakuo Takahashi; Toshiji Iwasaka

Angiotensin II (Ang II) is a potent vasopressor peptide that interacts with 2 major receptor isoforms - AT1 and AT2. Although blood pressure is increased in AT2 knockout mice, the underlying mechanisms remain undefined because of the low levels of expression of AT2 in the vasculature. Here we overexpressed AT2 in vascular smooth muscle (VSM) cells in transgenic (TG) mice. Aortic AT1 was not affected by overexpression of AT2. Chronic infusion of Ang II into AT2-TG mice completely abolished the AT1-mediated pressor effect, which was blocked by inhibitors of bradykinin type 2 receptor (icatibant) and nitric oxide (NO) synthase (L-NAME). Aortic explants from TG mice showed greatly increased cGMP production and diminished Ang II-induced vascular constriction. Removal of endothelium or treatment with icatibant and L-NAME abolished these AT2-mediated effects. AT2 blocked the amiloride-sensitive Na(+)/H(+) exchanger, promoting intracellular acidosis in VSM cells and activating kininogenases. The resulting enhancement of aortic kinin formation in TG mice was not affected by removal of endothelium. Our results suggest that AT2 in aortic VSM cells stimulates the production of bradykinin, which stimulates the NO/cGMP system in a paracrine manner to promote vasodilation. Selective stimulation of AT2 in the presence of AT1 antagonists is predicted to have a beneficial clinical effect in controlling blood pressure.


Circulation Research | 2003

Bone Marrow Monocyte Lineage Cells Adhere on Injured Endothelium in a Monocyte Chemoattractant Protein-1–Dependent Manner and Accelerate Reendothelialization as Endothelial Progenitor Cells

Soichiro Fujiyama; Katsuya Amano; Kazutaka Uehira; Masayuki Yoshida; Yasunobu Nishiwaki; Yoshihisa Nozawa; Denan Jin; Shinji Takai; Mizuo Miyazaki; Kensuke Egashira; Takayuki Imada; Toshiji Iwasaka; Hiroaki Matsubara

Abstract— Peripheral blood (PB)-derived CD14+ monocytes were shown to transdifferentiate into endothelial cell (EC) lineage cells and contribute to neovascularization. We investigated whether bone marrow (BM)- or PB-derived CD34−/CD14+ cells are involved in reendothelialization after carotid balloon injury. Although neither hematopoietic nor mesenchymal stem cells were included in human BM-derived CD34−/CD14+ monocyte lineage cells (BM-MLCs), they expressed EC-specific markers (Tie2, CD31, VE-cadherin, and endoglin) to an extent identical to mature ECs. When BM-MLCs were cultured with vascular endothelial growth factors, hematopoietic markers were drastically decreased and new EC-specific markers (Flk and CD34) were induced. BM-MLCs were intra-arterially transplanted into balloon-injured arteries of athymic nude rats. When BM-MLCs were activated by monocyte chemoattractant protein-1 (MCP-1) in vivo or in vitro, they adhered onto injured endothelium, differentiated into EC-like cells by losing hematopoietic markers, and inhibited neointimal hyperplasia. Ability to prevent neointimal hyperplasia was more efficient than that of BM-derived CD34+ cells. MCP-dependent adhesion was not observed in PB-derived CD34−/CD14+ monocytes. Regenerated endothelium exhibited a cobblestone appearance, blocked extravasation of dye, and induced NO-dependent vasorelaxation. Basal adhesive activities on HUVECs under laminar flow and &bgr;1-integrin expression (basal and active forms) were significantly increased in BM-MLCs compared with PB-derived monocytes. MCP-1 markedly enhanced adhesive activity of BM-MLCs (2.8-fold) on HUVECs by activating &bgr;1-integrin conformation. Thus, BM-MLCs can function as EC progenitors that are more potent than CD34+ cells and acquire the ability to adhere on injured endothelium in a MCP-1–dependent manner, leading to reendothelialization associated with inhibition of intimal hyperplasia. This will open a novel window to MCP-1–mediated biological actions and vascular regeneration strategies by cell therapy.


Circulation | 2002

Angiogenesis by Implantation of Peripheral Blood Mononuclear Cells and Platelets Into Ischemic Limbs

Osamu Iba; Hiroaki Matsubara; Yoshihisa Nozawa; Soichiro Fujiyama; Katsuya Amano; Yasukiyo Mori; Hiroyuki Kojima; Toshiji Iwasaka

Background—Peripheral blood mononuclear cells (PBMNCs), platelets, and polymorphonuclear leukocytes (PMNs) contain various angiogenic factors and cytokines. Methods and Results—Unilateral hindlimb ischemia was surgically induced in athymic nude rats, and fluorescence-labeled human blood cells (PBMNCs [107 cells]+platelets [109] or PBMNCs [107]+platelets [109]+PMNs [107]) were intramuscularly implanted into the ischemic limbs. Laser Doppler imaging revealed markedly increased blood perfusion in PBMNC+platelet-implanted limbs (44% increase, P <0.001) compared with control implantation of human umbilical vein vascular endothelial cells. The addition of PMNs to PBMNCs+platelets attenuated blood perfusion (27% decrease, P <0.01). Neocapillary densities were increased by implantation of PBMNCs+platelets or platelets alone (3.5-fold and 2.4-fold, respectively;P <0.001), whereas PMNs inhibited (32%, P <0.05) PBMNC+ platelet-mediated capillary formation. There was no incorporation of implanted PBMNCs into neocapillaries, whereas PBMNCs and platelets accumulated around arterioles after implantation. Cellular extract from PBMNCs+platelets, in which vascular endothelial growth factor (VEGF), basic fibroblast growth factor, platelet-derived growth factor-AB, and transforming growth factor-&bgr; were detected, markedly stimulated tubule formation of human umbilical vein vascular endothelial cells. Anti-VEGF neutralizing antibody markedly inhibited tubule formation and in vivo vessel formation. Neutrophil elastase inhibitor blocked the antiangiogenic action of PMNs, whereas inhibitors of oxygen metabolites had no effect. Conclusions—This study demonstrated that implantation of PBMNCs and platelets into ischemic limbs effectively induces collateral vessel formation by supplying angiogenic factors (mainly VEGF) and cytokines, suggesting that this cell therapy is useful as a novel strategy for therapeutic angiogenesis.


Circulation Research | 2001

Angiotensin AT1 and AT2 Receptors Differentially Regulate Angiopoietin-2 and Vascular Endothelial Growth Factor Expression and Angiogenesis by Modulating Heparin Binding–Epidermal Growth Factor (EGF)–Mediated EGF Receptor Transactivation

Soichiro Fujiyama; Hiroaki Matsubara; Yoshihisa Nozawa; Katsuya Maruyama; Yasukiyo Mori; Yoshiaki Tsutsumi; Hiroya Masaki; Yoko Uchiyama; Yoko Koyama; Atsuko Nose; Osamu Iba; Eriko Tateishi; Nahoko Ogata; Nobuo Jyo; Shigeki Higashiyama; Toshiji Iwasaka

Angiotensin II (Ang II)-mediated signals are transmitted via heparin binding epidermal growth factor (EGF)-like growth factor (HB-EGF) release followed by transactivation of EGF receptor (EGFR). Although Ang II and HB-EGF induce angiogenesis, their link to the angiopoietin (Ang)-Tie2 system remains undefined. We tested the effects of Ang II on Ang1, Ang2, or Tie2 expression in cardiac microvascular endothelial cells expressing the Ang II receptors AT(1) and AT(2). Ang II significantly induced Ang2 mRNA accumulations without affecting Ang1 or Tie2 expression, which was inhibited by protein kinase C inhibitors and by intracellular Ca(2+) chelating agents. Ang II transactivated EGFR via AT(1), and inhibition of EGFR abolished the induction of Ang2. Ang II caused processing of pro-HB-EGF in a metalloproteinase-dependent manner to stimulate maturation and release of HB-EGF. Neutralizing anti-HB-EGF antibody blocked EGFR phosphorylation by Ang II. Ang II also upregulated vascular endothelial growth factor (VEGF) expression in an HB-EGF/EGFR-dependent manner. AT(2) inhibited AT(1)-mediated Ang2 expression and phosphorylation of EGFR. In an in vivo corneal assay, AT(1) induced angiogenesis in an HB-EGF-dependent manner and enhanced the angiogenic activity of VEGF. Although neither Ang2 nor Ang1 alone induced angiogenesis, soluble Tie2-Fc that binds to angiopoietins attenuated AT(1)-mediated angiogenesis. These findings suggested that (1) Ang II induces Ang2 and VEGF expression without affecting Ang1 or Tie2 and (2) AT(1) stimulates processing of pro-HB-EGF by metalloproteinases, and the released HB-EGF transactivates EGFR to induce angiogenesis via the combined effect of Ang2 and VEGF, whereas AT(2) attenuates them by blocking EGFR phosphorylation. Thus, Ang II is involved in the VEGF-Ang-Tie2 system via HB-EGF-mediated EGFR transactivation, and this link should be considerable in pathological conditions in which collateral blood flow is required.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2002

Improvement of Collateral Perfusion and Regional Function by Implantation of Peripheral Blood Mononuclear Cells Into Ischemic Hibernating Myocardium

Hiroshi Kamihata; Hiroaki Matsubara; Takashi Nishiue; Soichiro Fujiyama; Katsuya Amano; Osamu Iba; Takanobu Imada; Toshiji Iwasaka

Objective—This study was performed to evaluate the angiogenic effect of implantation of peripheral blood mononuclear cells (PB-MNCs) compared with bone marrow mononuclear cells (BM-MNCs) into ischemic hibernating myocardium. Methods and Results—A NOGA electromechanical system was used to map the hibernating region and to inject cells. PB-MNCs and BM-MNCs contained similar levels of vascular endothelial growth factor and basic fibroblast growth factor, whereas contents of angiogenic cytokines (interleukin-1&bgr; and tumor necrosis factor-&agr;) were larger in PB-MNCs. Numbers of endothelial progenitors were ≈500-fold higher in BM-MNCs. In BM-MNC–implanted myocardia of pigs, an increase in systolic function (ejection fraction from 33% to 52%) and regional blood flow (2.1-fold) and a reduction of the ischemic area (from 29% to 8%) were observed. PB-MNC implantation reduced the ischemic area (from 31% to 17%), the extent of which was less than that seen with BM-MNCs. In saline-implanted myocardium, the ischemic area expanded (from 28% to 38%), and systolic function deteriorated. Angiography revealed an increase in collateral vessel formation by PB-MNC or BM-MNC implantation. Capillary numbers were increased 2.6- and 1.7-fold by BM-MNC and PB-MNC implantation, respectively. BM-MNCs but not PB-MNCs were incorporated into neocapillaries. Conclusions—Catheter-based implantation of PB-MNCs can effectively improve collateral perfusion and regional function in hibernating ischemic myocardium by its ability to mainly supply angiogenic factors and cytokines.


Hypertension | 2003

Enhancement of ischemia-induced angiogenesis by eNOS overexpression

Katsuya Amano; Hiroaki Matsubara; Osamu Iba; Mitsuhiko Okigaki; Soichiro Fujiyama; Takanobu Imada; Hiroyuki Kojima; Yoshihisa Nozawa; Seinosuke Kawashima; Mitsuhiro Yokoyama; Toshiji Iwasaka

Abstract—It remains undetermined whether continuous endothelial nitric oxide (NO) overexpression exerts angiogenic action. We surgically induced hindlimb ischemia in transgenic mice overexpressing endothelial NO synthase in the endothelium (eNOS-Tg) and studied neocapillary formation, ischemia-induced vascular endothelial growth factor (VEGF) expression, cGMP accumulation, and Akt/PKB signaling. Laser Doppler imaging revealed a markedly increased recovery of blood perfusion in ischemic limbs of eNOS-Tg mice (44% increase) compared with that in wild-type mice. Angiography showed a marked increase in basal and ischemia-induced collateral vessel formation in eNOS-Tg mice. Basal capillary densities and tissue cGMP levels were increased in eNOS-Tg mice (1.8-fold and 1.6-fold versus wild-type mice, respectively). Ischemia-induced neocapillary formation and cGMP accumulation were markedly increased in eNOS-Tg mice (3.6-fold and 4.1-fold versus preischemia levels, respectively), whereas those in wild-type mice were much less (1.8-fold and 1.5-fold, respectively). Basal and time-dependent VEGF expression in ischemic muscles did not differ between eNOS-Tg and wild-type mice. Basal and VEGF-mediated Akt phosphorylation in aortas was similar between eNOS-Tg and wild-type mice. Aortic basal eNOS expression was increased 3.3-fold, and VEGF-mediated eNOS phosphorylation was markedly induced in aortas of eNOS-Tg compared with preischemia levels (4.2-fold), whereas much smaller changes were observed in wild-type mice (1.8-fold increase). Our study demonstrates that overexpression of eNOS protein causes a marked increase in neocapillary formation in response to tissue ischemia without affecting ischemia-induced VEGF expression or VEGF-mediated Akt phosphorylation.


Hypertension | 2001

Angiotensin II type 2 receptor inhibits epidermal growth factor receptor transactivation by increasing association of SHP-1 tyrosine phosphatase

Yasunobu Shibasaki; Hiroaki Matsubara; Yoshihisa Nozawa; Yasukiyo Mori; Hiroya Masaki; Atsushi Kosaki; Yoshiaki Tsutsumi; Yoko Uchiyama; Soichiro Fujiyama; Atsuko Nose; Osamu Iba; Eriko Tateishi; Takamasa Hasegawa; Masatsugu Horiuchi; Clara Nahmias; Toshiji Iwasaka

Angiotensin (Ang) II has 2 major receptor isoforms, Ang type 1 (AT1) and Ang type (AT2). AT1 transphosphorylates epidermal growth factor receptor (EGFR) to activate extracellular signal–regulated kinase (ERK). Although AT2 was shown to inactivate ERK, the action of AT2 on EGFR activation remains undefined. Using AT2-overexpressing vascular smooth muscle cells from AT2 transgenic mice, we studied these undefined actions of AT2. Maximal ERK activity induced by Ang II was increased 1.9- and 2.2-fold by AT2 inhibition, which was abolished by orthovanadate but not okadaic acid or pertussis toxin. AT2 inhibited AT1-mediated EGFR tyrosine phosphorylation by 63%. The activity of SHP-1 tyrosine phosphatase was significantly upregulated 1 minute after AT2 stimulation and association of SHP-1 with EGFR was increased, whereas AT2 failed to tyrosine phosphorylate SHP-1. Stable overexpression of SHP-1–dominant negative mutant completely abolished AT2-mediated inhibition of EGFR and ERK activation. AT1-mediated c-fos mRNA accumulation was attenuated by 48% by AT2 stimulation. Induction of fibronectin gene containing an AP-1 responsive element in its 5′-flanking region was decreased by 37% after AT2 stimulation, corresponding to the results of gel mobility assay with the AP-1 sequence of fibronectin as a probe. These findings suggested that AT2 inhibits ERK activity by inducing SHP-1 activity, leading to decreases in AP-1 activity and AP-1–regulated gene expression, in which EGFR dephosphorylation plays an important role via association of SHP-1.


Circulation | 2001

Implantation of Bone Marrow Mononuclear Cells Into Ischemic Myocardium Enhances Collateral Perfusion and Regional Function via Side Supply of Angioblasts, Angiogenic Ligands, and Cytokines

Hiroshi Kamihata; Hiroaki Matsubara; Takashi Nishiue; Soichiro Fujiyama; Yoshiaki Tsutsumi; Ryozo Ozono; Hiroya Masaki; Yasukiyo Mori; Osamu Iba; Eriko Tateishi; Atsushi Kosaki; Satoshi Shintani; Toyoaki Murohara; Tsutomu Imaizumi; Toshiji Iwasaka


Kidney International | 2001

Angiotensin II signaling and HB-EGF shedding via metalloproteinase in glomerular mesangial cells

Yoko Uchiyama-Tanaka; Hiroaki Matsubara; Yoshihisa Nozawa; Satoshi Murasawa; Yasukiyo Mori; Atsushi Kosaki; Katsuya Maruyama; Hiroya Masaki; Yasunobu Shibasaki; Soichiro Fujiyama; Atsuko Nose; Osamu Iba; Takamasa Hasagawa; Eriko Tateishi; Shigeki Higashiyama; Toshiji Iwasaka


Journal of Molecular and Cellular Cardiology | 2004

Mechanism for IL-1β-mediated neovascularization unmasked by IL-1β knock-out mice

Katsuya Amano; Mitsuhiko Okigaki; Yasushi Adachi; Soichiro Fujiyama; Yasukio Mori; Atsushi Kosaki; Toshiji Iwasaka; Hiroaki Matsubara

Collaboration


Dive into the Soichiro Fujiyama's collaboration.

Top Co-Authors

Avatar

Hiroaki Matsubara

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Toshiji Iwasaka

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar

Osamu Iba

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar

Katsuya Amano

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar

Yasukiyo Mori

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar

Hiroya Masaki

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mitsuhiko Okigaki

Kyoto Prefectural University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge