Network


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

Hotspot


Dive into the research topics where Yoko Oyama is active.

Publication


Featured researches published by Yoko Oyama.


Biochemical and Biophysical Research Communications | 2009

Edaravone attenuates cerebral ischemic injury by suppressing aquaporin-4

Kiyoshi Kikuchi; Salunya Tancharoen; Fumiyo Matsuda; Kamal Krishna Biswas; Takashi Ito; Yoko Morimoto; Yoko Oyama; Kazunori Takenouchi; Naoki Miura; Noboru Arimura; Yuko Nawa; Xiaojie Meng; Binita Shrestha; Shinichiro Arimura; Masahiro Iwata; Kentaro Mera; Hisayo Sameshima; Yoshiko Ohno; Ryuichi Maenosono; Yutaka Tajima; Terukazu Kuramoto; Kenji Nakayama; Minoru Shigemori; Yoshihiro Yoshida; Teruto Hashiguchi; Ikuro Maruyama; Ko-ichi Kawahara

Aquaporin-4 (AQP4) plays a role in the generation of post-ischemic edema. Pharmacological modulation of AQP4 function may thus provide a novel therapeutic strategy for the treatment of stroke, tumor-associated edema, epilepsy, traumatic brain injury, and other disorders of the central nervous system (CNS) associated with altered brain water balance. Edaravone, a free radical scavenger, is used for the treatment of acute ischemic stroke (AIS) in Japan. In this study, edaravone significantly reduced the infarct area and improved the neurological deficit scores at 24h after reperfusion in a rat transient focal ischemia model. Furthermore, edaravone markedly reduced AQP4 immunoreactivity and protein levels in the cerebral infarct area. In light of observations that edaravone specifically inhibited AQP4 in a rat transient focal ischemia model, we propose that edaravone might reduce cerebral edema through the inhibition of AQP4 expression following cerebral infarction.


Journal of Immunology | 2010

B Cell-Derived Vascular Endothelial Growth Factor A Promotes Lymphangiogenesis and High Endothelial Venule Expansion in Lymph Nodes

Binita Shrestha; Teruto Hashiguchi; Takashi Ito; Naoki Miura; Kazunori Takenouchi; Yoko Oyama; Ko-ichi Kawahara; Salunya Tancharoen; Yuya Kii; Noboru Arimura; Narimasa Yoshinaga; Satoshi Noma; Chandan Shrestha; Takao Nitanda; Shinichi Kitajima; Kimiyoshi Arimura; Masahiro Sato; Taiji Sakamoto; Ikuro Maruyama

Vascular endothelial growth factor A (VEGF-A) is a prominent growth factor for both angiogenesis and lymphangiogenesis. Recent studies have shown the importance of VEGF-A in enhancing the growth of lymphatic endothelial cells in lymph nodes (LNs) and the migration of dendritic cells into LNs. VEGF-A is produced in inflamed tissues and/or in draining LNs, where B cells are a possible source of this growth factor. To study the effect of B cell-derived VEGF-A, we created transgenic mice (CD19Cre/hVEGF-Afl) that express human VEGF-A specifically in B cells. We found that the human VEGF-A produced by B cells not only induced lymphangiogenesis in LNs, but also induced the expansion of LNs and the development of high endothelial venules. Contrary to our expectation, we observed a significant decrease in the Ag-specific Ab production postimmunization with OVA and in the proinflammatory cytokine production postinoculation with LPS in these mice. Our findings suggest immunomodulatory effects of VEGF-A: B cell-derived VEGF-A promotes both lymphangiogenesis and angiogenesis within LNs, but then suppresses certain aspects of the ensuing immune responses.


Cardiovascular Pathology | 2008

C-reactive protein induces high-mobility group box-1 protein release through activation of p38MAPK in macrophage RAW264.7 cells

Ko-ichi Kawahara; Kamal Krishna Biswas; Masako Unoshima; Takashi Ito; Kiyoshi Kikuchi; Yoko Morimoto; Masahiro Iwata; Salunya Tancharoen; Yoko Oyama; Kazunori Takenouchi; Yuko Nawa; Noboru Arimura; Meng Xiao Jie; Binita Shrestha; Naoki Miura; Toshiaki Shimizu; Kentaro Mera; Shinichiro Arimura; Noboru Taniguchi; Hideo Iwasaka; Sonshin Takao; Teruto Hashiguchi; Ikuro Maruyama

BACKGROUND C-reactive protein (CRP) is widely used as a sensitive biomarker for inflammation. Increasing evidence suggests that CRP plays a role in inflammation. High-mobility group box-1 (HMGB1), a primarily nuclear protein, is passively released into the extracellular milieu by necrotic or damaged cells and is actively secreted by monocytes/macrophages. Extracellular HMGB1 as a potent inflammatory mediator has stimulated immense curiosity in the field of inflammation research. However, the molecular dialogue implicated between CRP and HMGB1 in delayed inflammatory processes remains to be explored. METHODS AND RESULTS The levels of HMGB1 in culture supernatants were determined by Western blot analysis and enzyme-linked immunosorbent assay in macrophage RAW264.7 cells. Purified CRP induced the release of HMGB1 in a dose- and time-dependent fashion. Immunofluorescence analysis revealed nuclear translocation of HMGB1 in response to CRP. The binding of CRP to the Fc gamma receptor in RAW264.7 cells was confirmed by fluorescence-activated cell sorter analysis. Pretreatment of cells with IgG-Fc fragment, but not IgG-Fab fragment, efficiently blocked this binding. CRP triggered the activation of p38MAPK and ERK1/2, but not Jun N-terminal kinase. Moreover, both p38MAPK inhibitor SB203580 and small interfering RNA significantly suppressed the release of HMGB1, but not the MEK1/2 inhibitor U-0126. CONCLUSION We demonstrated for the first time that CRP, a prominent risk marker for inflammation including atherosclerosis, could induce the active release of HMGB1 by RAW264.7 cells through Fc gamma receptor/p38MAPK signaling pathways, thus implying that CRP plays a crucial role in the induction, amplification, and prolongation of inflammatory processes, including atherosclerotic lesions.


Journal of Pharmacology and Experimental Therapeutics | 2009

The Free Radical Scavenger Edaravone Rescues Rats from Cerebral Infarction by Attenuating the Release of High-Mobility Group Box-1 in Neuronal Cells

Kiyoshi Kikuchi; Ko-ichi Kawahara; Salunya Tancharoen; Fumiyo Matsuda; Yoko Morimoto; Takashi Ito; Kamal Krishna Biswas; Kazunori Takenouchi; Naoki Miura; Yoko Oyama; Yuko Nawa; Noboru Arimura; Masahiro Iwata; Yutaka Tajima; Terukazu Kuramoto; Kenji Nakayama; Minoru Shigemori; Yoshihiro Yoshida; Teruto Hashiguchi; Ikuro Maruyama

Edaravone, a potent free radical scavenger, is clinically used for the treatment of cerebral infarction in Japan. Here, we examined the effects of edaravone on the dynamics of high-mobility group box-1 (HMGB1), which is a key mediator of ischemic-induced brain damage, during a 48-h postischemia/reperfusion period in rats and in oxygen-glucose-deprived (OGD) PC12 cells. HMGB1 immunoreactivity was observed in both the cytoplasm and the periphery of cells in the cerebral infarction area 2 h after reperfusion. Intravenous administration of 3 and 6 mg/kg edaravone significantly inhibited nuclear translocation and HMGB1 release in the penumbra area and caused a 26.5 ± 10.4 and 43.8 ± 0.5% reduction, respectively, of the total infarct area at 24 h after reperfusion. Moreover, edaravone also decreased plasma HMGB1 levels. In vitro, edaravone dose-dependently (1–10 μM) suppressed OGD- and H2O2-induced HMGB1 release in PC12 cells. Furthermore, edaravone (3–30 μM) blocked HMGB1-triggered apoptosis in PC12 cells. Our findings suggest a novel neuroprotective mechanism for edaravone that abrogates the release of HMGB1.


Biochemical and Biophysical Research Communications | 2009

Minocycline attenuates both OGD-induced HMGB1 release and HMGB1-induced cell death in ischemic neuronal injury in PC12 cells.

Kiyoshi Kikuchi; Ko-ichi Kawahara; Kamal Krishna Biswas; Takashi Ito; Salunya Tancharoen; Yoko Morimoto; Fumiyo Matsuda; Yoko Oyama; Kazunori Takenouchi; Naoki Miura; Noboru Arimura; Yuko Nawa; Xiaojie Meng; Binita Shrestha; Shinichiro Arimura; Masahiro Iwata; Kentaro Mera; Hisayo Sameshima; Yoshiko Ohno; Ryuichi Maenosono; Yoshihiro Yoshida; Yutaka Tajima; Terukazu Kuramoto; Kenji Nakayama; Minoru Shigemori; Teruto Hashiguchi; Ikuro Maruyama

High mobility group box-1 (HMGB1), a non-histone DNA-binding protein, is massively released into the extracellular space from neuronal cells after ischemic insult and exacerbates brain tissue damage in rats. Minocycline is a semisynthetic second-generation tetracycline antibiotic which has recently been shown to be a promising neuroprotective agent. In this study, we found that minocycline inhibited HMGB1 release in oxygen-glucose deprivation (OGD)-treated PC12 cells and triggered the activation of p38mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinases (ERK1/2). The ERK kinase (MEK)1/2 inhibitor U-0126 and p38MAPK inhibitor SB203580 blocked HMGB1 release in response to OGD. Furthermore, HMGB1 triggered cell death in a dose-dependent fashion. Minocycline significantly rescued HMGB1-induced cell death in a dose-dependent manner. In light of recent observations as well as the good safety profile of minocycline in humans, we propose that minocycline might play a potent neuroprotective role through the inhibition of HMGB1-induced neuronal cell death in cerebral infarction.


Experimental and Therapeutic Medicine | 2013

Preventive effects of Morus alba L. anthocyanins on diabetes in Zucker diabetic fatty rats

Ariya Sarikaphuti; Thamthiwat Nararatwanchai; Teruto Hashiguchi; Takashi Ito; Sita Thaworanunta; Kiyoshi Kikuchi; Yoko Oyama; Ikuro Maruyama; Salunya Tancharoen

The mulberry plant (Morus alba L.) contains abundant anthocyanins (ANCs), which are natural antioxidants. The aim of this study was to determine the ANC composition of Thai Morus alba L. fruits and to assess the effect of an ANC extract on blood glucose and insulin levels in male leptin receptor-deficient Zucker diabetic fatty (ZDF) rats. The major components of the ANC extract were identified by high-performance liquid chromatography-electrospray ionization-mass spectrometry. ZDF and lean rats were treated with 125 or 250 mg ANCs/kg body weight, or 1% carboxymethylcellulose (CMC) twice daily for 5 weeks. Neither ANC dose had an effect on body weight. Following 5 weeks of treatment, glucose levels were observed to increase from 105.5±8.7 to 396.25±21 mg/dl (P<0.0001) in the CMC-treated ZDF rats; however, the glucose levels were significantly lower in the rats treated with 125 or 250 mg/kg ANCs (228.25±45 and 131.75±10 mg/dl, respectively; P<0.001 versus CMC). The administration of 250 mg/kg ANCs normalized glucose levels in the ZDF rats towards those of the lean littermates. Insulin levels were decreased significantly in the ZDF rats treated with CMC or 125 mg/kg ANCs (P<0.0001), but not in the rats treated with 250 mg/kg ANCs. Histologically, 250 mg/kg ANCs was observed to prevent islet degeneration compared with the islets in CMC-treated rats. This study, demonstrated that ANCs extracted from Morus alba L. were well tolerated and exhibited effective anti-diabetic properties in ZDF rats. ANCs represent a promising class of therapeutic compounds that may be useful in the prevention of type 2 diabetes.


PLOS ONE | 2014

HMGB1 Promotes the Development of Pulmonary Arterial Hypertension in Rats

Yukari Sadamura-Takenaka; Takashi Ito; Satoshi Noma; Yoko Oyama; Shingo Yamada; Ko-ichi Kawahara; Hiromasa Inoue; Ikuro Maruyama

Rationale Pulmonary arterial hypertension (PAH) is characterized by increased pulmonary vascular resistance leading to right ventricular failure and death. Recent studies have suggested that chronic inflammatory processes are involved in the pathogenesis of PAH. However, the molecular and cellular mechanisms driving inflammation have not been fully elucidated. Objectives To elucidate the roles of high mobility group box 1 protein (HMGB1), a ubiquitous DNA-binding protein with extracellular pro-inflammatory activity, in a rat model of PAH. Methods Male Sprague-Dawley rats were administered monocrotaline (MCT). Concentrations of HMGB1 in bronchoalveolar lavage fluid (BALF) and serum, and localization of HMGB1 in the lung were examined over time. The protective effects of anti-HMGB1 neutralizing antibody against MCT-induced PAH were tested. Results HMGB1 levels in BALF were elevated 1 week after MCT injection, and this elevation preceded increases of other pro-inflammatory cytokines, such as TNF-α, and the development of PAH. In contrast, serum HMGB1 levels were elevated 4 weeks after MCT injection, at which time the rats began to die. Immunohistochemical analyses indicated that HMGB1 was translocated to the extranuclear space in periarterial infiltrating cells, alveolar macrophages, and bronchial epithelial cells of MCT-injected rats. Anti-HMGB1 neutralizing antibody protected rats against MCT-induced lung inflammation, thickening of the pulmonary artery wall, and elevation of right ventricular systolic pressure, and significantly improved the survival of the MCT-induced PAH rats. Conclusions Our results identify extracellular HMGB1 as a promoting factor for MCT-induced PAH. The blockade of HMGB1 activity improved survival of MCT-induced PAH rats, and thus might be a promising therapy for the treatment of PAH.


Medical Hypotheses | 2010

Edaravone: a new therapeutic approach for the treatment of acute stroke.

Kiyoshi Kikuchi; Ko-ichi Kawahara; Naohisa Miyagi; Terukazu Kuramoto; Yoko Morimoto; Salunya Tancharoen; Naoki Miura; Kazunori Takenouchi; Yoko Oyama; Binita Shrestha; Fumiyo Matsuda; Yoshihiro Yoshida; Shinichiro Arimura; Kentaro Mera; Ko-ichi Tada; Narimasa Yoshinaga; Ryuichi Maenosono; Yoshiko Ohno; Teruto Hashiguchi; Ikuro Maruyama; Minoru Shigemori

Acute stroke, including acute ischemic stroke (AIS) and acute hemorrhagic stroke, (AHS) is a common medical problem with particular relevance to the demographic changes in industrialized societies. In recent years, treatments for AIS have emerged, including thrombolysis with tissue plasminogen activator (t-PA). Although t-PA is the most effective currently available therapy, it is limited by a narrow therapeutic time window and side effects, and only 3% of all AIS patients receive thrombolysis. Edaravone was originally developed as a potent free radical scavenger and, since 2001, has been widely used to treat AIS in Japan. It was shown that edaravone extended the narrow therapeutic time window of t-PA in rats. The therapeutic time window is very important for the treatment of AIS, and early edaravone treatment is more effective. Thus, more AIS patients might be rescued by administering edaravone with t-PA. Meanwhile, edaravone attenuates AHS-induced brain edema, neurologic deficits and oxidative injury in rats. Although edaravone treatment is currently only indicated for AIS, it does offer neuroprotective effects against AHS in rats. Therefore, we hypothesize that early administration of edaravone can rescue AHS patients as well as AIS patients. Taken together, our findings suggest that edaravone should be immediately administered on suspicion of acute stroke, including AIS and AHS.


Biochemical and Biophysical Research Communications | 2017

Low grade inflammation inhibits VEGF induced HUVECs migration in p53 dependent manner

Sushil Panta; Munekazu Yamakuchi; Toshiaki Shimizu; Kazunori Takenouchi; Yoko Oyama; Toyoyasu Koriyama; Tsuyoshi Kojo; Teruto Hashiguchi

In the course of studying crosstalk between inflammation and angiogenesis, high doses of pro-inflammatory factors have been reported to induce apoptosis in cells. Under normal circumstances also the pro-inflammatory cytokines are being released in low doses and are actively involved in cell signaling pathways. We studied the effects of low grade inflammation in growth factor induced angiogenesis using tumor necrosis factor alfa (TNFα) and vascular endothelial growth factor A (VEGF) respectively. We found that low dose of TNFα can inhibit VEGF induced angiogenesis in human umbilical vein endothelial cells (HUVECs). Low dose of TNFα induces mild upregulation and moreover nuclear localization of tumor suppressor protein 53 (P53) which causes decrease in inhibitor of DNA binding-1 (Id1) expression and shuttling to the cytoplasm. In absence of Id1, HUVECs fail to upregulate β3-integrin and cell migration is decreased. Connecting low dose of TNFα induced p53 to β3-integrin through Id1, we present additional link in cross talk between inflammation and angiogenesis.


Therapeutic Apheresis and Dialysis | 2017

Decreased Expression of Thrombomodulin in Endothelial Cells by Fibroblast Growth Factor-23/α-Klotho: FGF23/α-Klotho Downregulation of Thrombomodulin

Kenji F. Tanaka; Tancharoen Salunya; Yoshihiro Motomiya; Yasuki Motomiya; Yoko Oyama; Munekazu Yamakuchi; Ikuro Maruyama

Chronic kidney disease (CKD) has been known to be a state of excessive fibroblast growth factor‐23 (FGF23) and α‐Klotho deficiency. Patients undergoing hemodialysis have an increased mortality risk associated with cardiovascular disease and endothelial dysfunction. The mechanism responsible for the relationship of FGF23 to endothelial damage in these patients has been unclear. On the other hands, increasing evidences have demonstrated that thrombomodulin (TM) plays an important role in the endothelial barrier. Here, we report the suppression of membrane TM, in a dose‐dependent manner, in human umbilical vein endothelial cells after FGF23 and FGF23/α‐Klotho stimulation. In addition, the levels of soluble TM, which reflect endothelial cell injury, were much higher in cell supernatants after FGF23 and FGF23/α‐Klotho stimulation than in the control supernatant. This study indicates a possible mechanism by which excessive levels of FGF23 are involved in endothelial TM disruption, which has been implicated as a potential cardiovascular risk factor in patients with CKD, especially in HD patients.

Collaboration


Dive into the Yoko Oyama's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ko-ichi Kawahara

Osaka Institute of Technology

View shared research outputs
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