Network


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

Hotspot


Dive into the research topics where Hideki Amano is active.

Publication


Featured researches published by Hideki Amano.


Journal of Experimental Medicine | 2003

Host Prostaglandin E2-EP3 Signaling Regulates Tumor-Associated Angiogenesis and Tumor Growth

Hideki Amano; Izumi Hayashi; Hirahito Endo; Hidero Kitasato; Shohei Yamashina; Takayuki Maruyama; Michiyoshi Kobayashi; Kazutoyo Satoh; Masami Narita; Yukihiko Sugimoto; Takahiko Murata; Hirokuni Yoshimura; Shuh Narumiya; Masataka Majima

Nonsteroidal antiinflammatories are known to suppress incidence and progression of malignancies including colorectal cancers. However, the precise mechanism of this action remains unknown. Using prostaglandin (PG) receptor knockout mice, we have evaluated a role of PGs in tumor-associated angiogenesis and tumor growth, and identified PG receptors involved. Sarcoma-180 cells implanted in wild-type (WT) mice formed a tumor with extensive angiogenesis, which was greatly suppressed by specific inhibitors for cyclooxygenase (COX)-2 but not for COX-1. Angiogenesis in sponge implantation model, which can mimic tumor-stromal angiogenesis, was markedly suppressed in mice lacking EP3 (EP3−/−) with reduced expression of vascular endothelial growth factor (VEGF) around the sponge implants. Further, implanted tumor growth (sarcoma-180, Lewis lung carcinoma) was markedly suppressed in EP3−/−, in which tumor-associated angiogenesis was also reduced. Immunohistochemical analysis revealed that major VEGF-expressing cells in the stroma were CD3/Mac-1 double-negative fibroblasts, and that VEGF-expression in the stroma was markedly reduced in EP3−/−, compared with WT. Application of an EP3 receptor antagonist inhibited tumor growth and angiogenesis in WT, but not in EP3−/−. These results demonstrate significance of host stromal PGE2-EP3 receptor signaling in tumor development and angiogenesis. An EP3 receptor antagonist may be a candidate of chemopreventive agents effective for malignant tumors.


Laboratory Investigation | 2003

COX-2/VEGF-dependent facilitation of tumor-associated angiogenesis and tumor growth in vivo.

Satoko Yoshida; Hideki Amano; Izumi Hayashi; Hidero Kitasato; Mariko Kamata; Madoka Inukai; Hirokuni Yoshimura; Masataka Majima

Nonsteroidal anti-inflammatory drugs are known to suppress the occurrence and progression of malignancies such as colorectal cancers. However, the precise mechanism of these actions remains unknown. We have evaluated the role of an inducible cyclo-oxygenase (COX-2) in tumor-associated angiogenesis and tumor growth, and identified the downstream molecules involved using a ddy mouse model of sponge angiogenesis, which mimics tumor angiogenesis and is COX-2 and vascular endothelial growth factor (VEGF) dependent. In this model, VEGF expression was down-regulated by selective COX-2 inhibition with NS-398. To find out the involvement of COX-2/VEGF pathway in tumor-associated angiogenesis, we estimated angiogenesis occurring around implanted Millipore chambers containing sarcoma-180 (S-180) cells or Lewis lung carcinoma cells. Daily oral administration of NS-398 or of aspirin, a nonselective COX inhibitor, suppressed angiogenesis seen around the Millipore chambers. S-180 cells implanted in ddy mice formed substantial tumors with extensive angiogenesis markedly suppressed by aspirin and COX-2 inhibitors NS-398 and JTE522, but not by mofezolac, an inhibitor of constitutive COX-1. Tumor-associated angiogenesis was also significantly suppressed by a neutralizing antibody against VEGF. S-180 tumor growth in the subcutaneous tissues was also suppressed by aspirin, COX-2 selective inhibitors, and the VEGF antibody, but not by the COX-1 inhibitor. These results demonstrate that the inhibition of the COX-2/VEGF-dependent pathway was effective in tumor-associated angiogenesis, tumor growth, and tumor metastasis.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Neuronal system-dependent facilitation of tumor angiogenesis and tumor growth by calcitonin gene-related peptide

Masaya Toda; Tatsunori Suzuki; Kanako Hosono; Izumi Hayashi; Shinichiro Hashiba; Yuichiro Onuma; Hideki Amano; Yukiko Kurihara; Hiroki Kurihara; Hirotsugu Okamoto; Sumio Hoka; Masataka Majima

A neuropeptide, calcitonin gene-related peptide (CGRP), is widely distributed in neuronal systems and exhibits numerous biological activities. Using CGRP-knockout mice (CGRP−/−), we examined whether or not endogenous CGRP facilitates angiogenesis indispensable to tumor growth. CGRP increased tube formation by endothelial cells in vitro and enhanced sponge-induced angiogenesis in vivo. Tumor growth and tumor-associated angiogenesis in CGRP−/− implanted with Lewis lung carcinoma (LLC) cells were significantly reduced compared with those in wild-type (WT) mice. A CGRP antagonist, CGRP8-37 or denervation of sciatic nerves (L1–5) suppressed LLC growth in the sites of denervation compared with vehicle infusion or sham operation. CGRP precursor mRNA levels in the dorsal root ganglion in LLC-bearing WT were increased compared with those in non-LLC-bearing mice. This increase was abolished by denervation. The expression of VEGF in tumor stroma was down-regulated in CGRP−/−. These results indicate that endogenous CGRP facilitates tumor-associated angiogenesis and tumor growth and suggest that relevant CGRP may be derived from neuronal systems including primary sensory neurons and may become a therapeutic target for cancers.


Laboratory Investigation | 2002

Suppressed angiogenesis in kininogen-deficiencies.

Izumi Hayashi; Hideki Amano; Satoko Yoshida; Kazuhisa Kamata; Mariko Kamata; Madoka Inukai; Tomoe Fujita; Yuji Kumagai; Sen-ichi Furudate; Masataka Majima

We investigated whether the kinin-generating system enhanced angiogenesis in chronic and proliferative granuloma and in tumor-surrounding stroma. In rat sponge implants, angiogenesis was gradually developed in normal Brown Norway Kitasato rats (BN-Ki). The development of angiogenesis was significantly suppressed in kininogen-deficient Brown Norway Katholiek rats (BN-Ka). The angiogenesis enhanced by basic fibroblast growth factor was also significantly less marked in BN-Ka than in BN-Ki. Naturally occurring angiogenesis was significantly suppressed by B1 or B2 antagonist. mRNA of vascular endothelial growth factor was more highly expressed in the granulation tissues in BN-Ki than in BN-Ka. Daily topical injections of aprotinin, but not of soy bean trypsin inhibitor, suppressed angiogenesis. Daily topical injections of low-molecular weight kininogen enhanced angiogenesis in BN-Ka. Topical injections of serum from BN-Ki, but not from BN-Ka, also facilitated angiogenesis in BN-Ka. FR190997, a nonpeptide mimic of bradykinin, promoted angiogenesis markedly, with concomitant increases in vascular endothelial growth factor mRNA. Angiogenesis in the granulation tissues around the implanted Millipore chambers containing Walker-256 cells was markedly more suppressed in BN-Ka than in BN-Ki. Our results suggest that endogenous kinin generated from the tissue kallikrein-kinin system enhances angiogenesis in chronic and proliferative granuloma and in the stroma surrounding a tumor. Thus, the agents for the kinin-generating system and/or kinin receptor signaling may become useful tools for controlling angiogenesis.


Journal of Clinical Investigation | 2014

Prostanoid induces premetastatic niche in regional lymph nodes

Fumihiro Ogawa; Hideki Amano; Koji Eshima; Yoshiya Ito; Yoshio Matsui; Kanako Hosono; Hidero Kitasato; Akira Iyoda; Kazuya Iwabuchi; Yuji Kumagai; Yukitoshi Satoh; Shuh Narumiya; Masataka Majima

The lymphatic system is an important route for cancer dissemination, and lymph node metastasis (LNM) serves as a critical prognostic determinant in cancer patients. We investigated the contribution of COX-2-derived prostaglandin E2 (PGE2) in the formation of a premetastatic niche and LNM. A murine model of Lewis lung carcinoma (LLC) cell metastasis revealed that COX-2 is expressed in DCs from the early stage in the lymph node subcapsular regions, and COX-2 inhibition markedly suppressed mediastinal LNM. Stromal cell-derived factor-1 (SDF-1) was elevated in DCs before LLC cell infiltration to the lymph nodes, and a COX-2 inhibitor, an SDF-1 antagonist, and a CXCR4 neutralizing antibody all reduced LNM. Moreover, LNM was reduced in mice lacking the PGE2 receptor EP3, and stimulation of cultured DCs with an EP3 agonist increased SDF-1 production. Compared with WT CD11c+ DCs, injection of EP3-deficient CD11c+ DCs dramatically reduced accumulation of SDF-1+CD11c+ DCs in regional LNs and LNM in LLC-injected mice. Accumulation of Tregs and lymph node lymphangiogenesis, which may influence the fate of metastasized tumor cells, was also COX-2/EP3-dependent. These results indicate that DCs induce a premetastatic niche during LNM via COX-2/EP3-dependent induction of SDF-1 and suggest that inhibition of this signaling axis may be an effective strategy to suppress premetastatic niche formation and LNM.


Biomedicine & Pharmacotherapy | 2010

Host prostaglandin EP3 receptor signaling relevant to tumor-associated lymphangiogenesis

Hidefumi Kubo; Kanako Hosono; Tatsunori Suzuki; Yasufumi Ogawa; Hiroshi Kato; Hiroki Kamata; Yoshiya Ito; Hideki Amano; Tetsuki Kato; Hiroyuki Sakagami; Izumi Hayashi; Yukihiko Sugimoto; Shuh Narumiya; Masahiko Watanabe; Masataka Majima

Prostaglandin E(2) (PGE(2)) and prostaglandin E (EP) receptor signaling pathways have been implicated in the promotion of tumor growth and angiogenesis. However, little is known about their roles in lymphangiogenesis during tumor development. The present study evaluates whether endogenous PGE(2) exhibits a critical role in tumor-associated lymphangiogenesis. Treatment of male C57BL/6 mice with a cyclooxygenase-2 inhibitor, celecoxib, for seven days resulted in a 52.4% reduction in tumor size induced by subcutaneous injection of murine Lewis lung cells. Celecoxib treatment down-regulated the expression of vascular endothelial growth factor receptor (VEGFR)-3 in stromal tissues by 73.9%, and attenuated expression of podoplanin, a marker for lymphatic endothelial cells. To examine the role of host PGE receptor signaling, we tested four kinds of EP receptor knockout mice. At Day 7 after tumor cell implantation, EP3 receptor knockout mice, but not EP receptor knockout mice lacking EP1, EP2, or EP4, exhibited a 53.3% reduction in tumor weight, which was associated with a 74.5% reduction in VEGFR-3 mRNA expression in tumor stromal tissues. At Day 14, VEGFR-3 expression in EP3-/- mice remained significantly lower than that of their wild-type (WT) counterparts. The expression of VEGF-C in the tumor stromal tissues in EP3-/- mice were also reduced by 22.1% (Day 7) and 44.1% (Day 14), respectively. In addition, the level of immunoreactive podoplanin in the tumor tissues from EP3-/- mice was less than that of WT. These results suggest that host EP3 receptor signaling regulates tumor-associated lymphangiogenesis by up-regulating expression of VEGF-C and its receptor, VEGFR-3, in tumor stromal tissues. Host EP3 blockade together with COX-2 inhibition may be a novel therapeutic strategy to suppress tumor-associated lymphangiogenesis.


Biomedicine & Pharmacotherapy | 2015

Suppressed recruitment of alternatively activated macrophages reduces TGF-β1 and impairs wound healing in streptozotocin-induced diabetic mice.

Shin-ichiro Okizaki; Yoshiya Ito; Kanako Hosono; Kazuhito Oba; Hirotoki Ohkubo; Hideki Amano; Masayoshi Shichiri; Masataka Majima

BACKGROUND Diabetes mellitus inhibits wound-induced angiogenesis, impairing the wound healing process and leading to the development of chronic wounds. Impaired healing of diabetic wounds is caused by persistent pro-inflammatory macrophages recruited to the granulation tissue; however, little is known about the phenotype of the macrophages involved in diabetic wound healing. The present study was conducted to examine the involvement of macrophages in impaired wound healing using streptozotocin (STZ)-induced diabetic mice. METHODS Full-thickness skin wounds were created on the backs of mice treated with STZ or vehicle. RESULTS Compared with controls, wound healing and angiogenesis were suppressed in STZ-treated mice, with attenuated expression of vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF)-2 in wound granulation tissue. STZ-treated mice exhibited enhanced recruitment of classically activated macrophages (M1) expressing inducible nitric oxide synthase (iNOS) and suppressed recruitment of alternatively activated macrophages (M2) expressing transforming growth factor-beta-1 (TGF-β1). Treatment of diabetic mice with TGF-β1 restored wound healing and angiogenesis and normalized M1/M2 macrophage polarization in the granulation tissue. CONCLUSIONS These results suggest that an imbalance of macrophage phenotypes contributes to impaired wound healing in STZ-induced diabetic mice, and treatment with cytokines derived from M2 macrophages may be an effective therapeutic strategy to increase angiogenesis and promote healing of diabetic wounds.


Cancer Science | 2012

Thromboxane A 2 receptor signaling facilitates tumor colonization through P-selectin-mediated interaction of tumor cells with platelets and endothelial cells

Yoshio Matsui; Hideki Amano; Yoshiya Ito; Koji Eshima; Tastunori Suzuki; Fumihiro Ogawa; Akira Iyoda; Yukitoshi Satoh; Shintaro Kato; Masaki Nakamura; Hidero Kitasato; Shuh Narumiya; Masataka Majima

Thromboxane A2 (TXA2) is a prostanoid formed by thromboxane synthase using the cyclooxygenase product, prostaglandin H(2), as the substrate. TXA2 was shown to enhance tumor metastasis, but the underlying mechanism remains unclear. B16F1 melanoma cells were intravenously injected into TXA2 receptor (TP) knockout mice (TP−/−) and wild‐type littermates (WT). TP−/− showed a reduction in B16F1 lung colonization and mortality rate, which were associated with a decreased number of platelets. Platelet activation as assessed by P‐selectin expression was suppressed in TP−/−. A selective P‐selectin neutralizing antibody decreased the lung colonization in WT mice, but not in TP−/−. The expression of P‐selectin glycoprotein ligand‐1 in B16F1 and HUVEC were enhanced by treatment with U46619, a thromboxane analog. The plasma levels of vascular endothelial growth factor (VEGF) and stromal‐derived factor (SDF)‐1 were lower in TP−/−. In TP−/−, the mobilization of progenitor cells expressing CXCR4+VEGFR1+ from bone marrow and the recruitment of those cells to lung tissues were suppressed. These results suggest that TP signaling plays a critical role in tumor colonization through P‐selectin‐mediated interactions between platelets‐tumor cells and tumor cells‐endothelial cells through the TP signaling‐dependent production of VEGF and SDF‐1, which might be involved in the mobilization of VEGFR1+CXCR4+ cells. Blockade of TP signaling might be useful in the treatment of tumor metastasis. (Cancer Sci 2012; 103: 700–707)


Human Cell | 2002

Cyclooxygenase-2 and adenylate cyclase/protein kinase A signaling pathway enhances angiogenesis through induction of vascular endothelial growth factor in rat sponge implants.

Hideki Amano; Izumi Haysahi; Satoko Yoshida; Hirokuni Yoshimura; Masataka Majima

Angiogenesis is reportedly enhanced by prostaglandins (PGs). In the present experiment, we tested whether or not COX-2 and adenylate cyclase/protein kinase A (AC/PKA)-dependent VEGF induction enhanced angiogenesis in this model. Angiogenesis was enhanced by topical injection of human recombinant basic fibroblast growth factor (bFGF). The enhanced angiogenesis by bFGF was inhibited by indomethacin or selective COX-2 inhibitors, NS398, nimesulide, and JTE-522. Topical daily injections of 8-bromo-cAMP enhanced angiogenesis in a dose-dependent manner. Forskolin. an activator of AC, also facilitated angiogenesis in a dose-dependent manner, as did amrinone, an inhibitor of phosphodiesterase. VEGF induction was confirmed by the increased levels in the fluids in the sponge matrix after topical injection of 8-bromo-CAMP. Immunohistochemical investigation further revealed the VEGF-expressed cells in the sponge granulation tissues to be fibroblasts, and the intensity of positive reactions was enhanced by bFGF, 8-bromo-cAMP. forskolin, and amrinone. Angiogenesis was inhibited by indometacin or selective COX-2 inhibitors, NS-398, nimesulide, and JTE-522. In addition, angiogenesis without topical injections of the above compounds was also suppressed by SQ22,536, an inhibitor for AC, or H-89, an inhibitor for PKA, with concomitant reductions in VEGF levels. Daily topical injections of neutralizing antibody or anti-sense oligonucleotide against VEGF significantly suppressed angiogenesis. These results suggested that COX-2 and AC/PKA-dependent induction of VEGF certainly enhanced angiogenesis, and that pharmacological tools for controlling this signaling pathway may be able to facilitate the management of conditions involving angiogenesis.


Biomedicine & Pharmacotherapy | 2009

Blockade of an angiotensin type I receptor enhances effects of radiation on tumor growth and tumor-associated angiogenesis by reducing vascular endothelial growth factor expression

Yuichiro Ohnuma; Masaya Toda; Mamoru Fujita; Kanako Hosono; Tatsunori Suzuki; Yasuhumi Ogawa; Hideki Amano; Hidero Kitasato; Kazushige Hayakawa; Masataka Majima

OBJECTIVE Angiogenesis, the formation of new capillary blood vessels, is essential for tumor progression. We had reported that Type 1 angiotensin receptor (AT1-R) antagonist reduced tumor-associated angiogenesis. Since antiangiogenic agents were reported to enhance efficacy of radiation therapy, we tested here whether or not AT1-R blockade facilitates the effects of radiation. METHODS 1 x 10(6) LLC cells were injected into the subcutaneous tissue of male C57BL/6 mice, and when the average tumor volume reached around 0.1 cm(3), radiation doses (3, 5, 10, and 15 Gy) were given on day 1. RESULTS The mean tumor volumes at day 22 were 6.39 (3 Gy), 6.15 (5 Gy), 5.15 (10 Gy), and 3.07 (15 Gy) cm(3), respectively. Combination of 10 Gy radiation with AT1R antagonist TCV-116 (30 mg/kg) significantly inhibited tumor growth by 83% (1.47 +/- 0.11 cm(3), P < 0.01) in comparison with its inhibition of control tumors (8.81 +/- 0.45 cm(3)). The same was true for mean vessel density, and the combination therapy markedly reduced tumor-associated angiogenesis. This was confirmed by the reduced expression of CD31. LLC tumor growth was blocked by neutralizing antibody against vascular endothelial growth factor (VEGF). Real-time PCR analysis of VEGF disclosed a marked reduction in the mice under combination therapy, compared with control mice. CONCLUSIONS These results suggest that combination of radiation with AT1-R blockade markedly reduced the LLC growth rate, and that this was due to reduction of neovascularization by reducing VEGF levels. Combination therapy consisting of radiation and AT1R blockade may become an effective novel strategy for cancer treatment.

Collaboration


Dive into the Hideki Amano'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