Network


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

Hotspot


Dive into the research topics where Yoko Morimoto is active.

Publication


Featured researches published by Yoko Morimoto.


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.


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

MK615 attenuates Porphyromonas gingivalis lipopolysaccharide-induced pro-inflammatory cytokine release via MAPK inactivation in murine macrophage-like RAW264.7 cells.

Yoko Morimoto; Kiyoshi Kikuchi; Takashi Ito; Masayuki Tokuda; Takashi Matsuyama; Satoshi Noma; Teruto Hashiguchi; Mitsuo Torii; Ikuro Maruyama; Ko-ichi Kawahara

The Japanese apricot, known as Ume in Japanese, has been a traditional Japanese medicine for centuries, and is a familiar and commonly consumed food. The health benefits of Ume are now being widely recognized and have been strengthened by recent studies showing that MK615, an extract of compounds from Ume, has strong anticancer and anti-inflammatory effects. However, the potential role of MK615 in the periodontal field remains unknown. Here, we found that MK615 significantly reduced the production of pro-inflammatory mediators (tumor necrosis factor-alpha and interleukin-6) induced by Porphyromonas gingivalis lipopolysaccharide (LPS), a major etiological agent in localized chronic periodontitis, in murine macrophage-like RAW264.7 cells. MK615 markedly inhibited the phosphorylation of ERK1/2, p38MAPK, and JNK, which is associated with pro-inflammatory mediator release pathways. Moreover, MK615 completely blocked LPS-triggered NF-kappaB activation. The present results suggest that MK615 has potential as a therapeutic agent for treating inflammatory diseases such as periodontitis.


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.


International Journal of Molecular Medicine | 2011

Beyond neurological disease: New targets for edaravone (Review)

Kiyoshi Kikuchi; Naohisa Miyagi; Yoko Morimoto; Takashi Ito; Salunya Tancharoen; Naoki Miura; Kei Miyata; Rokudai Sakamoto; Chiemi Kikuchi; Narumi Iida; Naoto Shiomi; Terukazu Kuramoto; Ko-ichi Kawahara

Free radicals play major roles in the pathogenesis of tissue damage in many diseases and clinical conditions, and the removal of free radicals may offer a treatment option. Several modulators of free radical scavenger pathways have been developed and some have progressed to clinical trials. One such agent, edaravone, was approved in 2001 in Japan for the treatment of cerebral infarction. It has since been shown that edaravone can diffuse into many organs and, in addition to its effects on hydroxyl radical removal, edaravone modulates inflammatory processes, matrix metalloproteinase levels, nitric oxide production, apoptotic cell death, and necrotic cell death. Edaravone also exerts protective effects in a number of animal models of disease and tissue damage, including models of myocardial, lung, intestinal, liver, pancreatic and renal injury. Together with the proven safety of edaravone following 9 years of use as a modulator of free radical scavenging pathways in neurological disease, these additional effects of edaravone suggest that it may offer a novel treatment for several non-neurological diseases and clinical conditions in humans.


Xenotransplantation | 2007

HMGB1 release in co-cultures of porcine endothelial and human T cells

Ko-ichi Kawahara; Kentaro Setoyama; Kiyoshi Kikuchi; Kamal Krishna Biswas; Ryozo Kamimura; Masahiro Iwata; Takashi Ito; Yoko Morimoto; Teruto Hashiguchi; Sonshin Takao; Ikuro Maruyama

Abstract:  High mobility group box‐1 (HMGB1) protein, primarily from the nucleus, is released into the extracellular milieu either passively by necrotic or damaged cells, or actively by secretion from monocytes/macrophages. Extracellular HMGB1 acts as a potent inflammatory stimulator by promoting cytokine (for example, tumor necrosis factor‐α) production, and also has pro‐coagulant activity. The signaling pathway initiated by receptor for advanced glycation end‐product (RAGE), which is the HMGB1 receptor, also induces complement activation. Recent studies have implicated HMGB1 in acute cardiac allograft rejection, and have identified infiltrating T cells and other damaged cells as its main sources. HMGB1 blockade using the anti‐HMGB1 antibody HMGB1 box‐A (amino‐terminal region) and soluble RAGE rescues mice from acute rejection. We therefore studied the release of HMGB1 in co‐cultures of porcine aortic endothelial cells (PAEC) and human leukocytes. Human T cells, but not B cells, monocytes or neutrophils, stimulated significant HMGB1 release in culture with PAEC; this activity required cell–cell contact and was dose‐dependent, as determined by Western blotting. The released HMGB1 originated from both cell types, as immunofluorescent microscopy showed that it was present in the cytosol of PAEC in contact with T cells, and had disappeared from the T‐cell nuclei. These results demonstrate that direct interactions between PAEC and T cells might be a key factor in triggering HMGB1 release, which suggests that HMGB1 is associated with graft rejection in the early phase.


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.


Journal of Neurology and Neurophysiology | 2011

Beneficial Effects of the Free Radical Scavenger Edaravone (Radicut) in Neurologic Diseases

Kiyoshi Kikuchi; Naoki Miura; Yoko Morimoto; Takashi Ito; Salunya Tancharoen; Kei Miyata; Chiemi Kikuchi; Narumi Iida; Nobuyuki Takeshige; Naohisa Miyagi; Naoto Shiomi; Terukazu Kuramoto; Teruto Hashiguchi; Ikuro Maruyama; Motohiro Morioka; Ko-ichi Kawahara

Free radicals play major roles in the pathogenesis of many diseases, including neurologic diseases, making them an attractive target for therapeutic intervention. Several free radical scavengers have been developed, and some have progressed to clinical trials for the treatment of ischemic stroke. One such scavenger, edaravone is currently used to treat patients who present within 24 h of an attack. Edaravone can diffuse into many affected organs. Edaravone also exerts protective effects against brain and spinal cord injuries. Beyond its direct free radical scavenging effect, edaravone has anti-apoptotic and anti-inflammatory effects in various diseases. Here, we critically review the literature on experimental animal model and clinical studies of edaravone efficacy, and examine whether it should be considered a candidate for worldwide development. Edaravone has proven safe during 10 years of use as a free radical scavenger to treat ischemic stroke. In addition to ischemic stroke treatment, animal data suggest that edaravone may be an effective treatment option for several neurologic diseases, but additional clinical trials are necessary to verify its efficacy.


Journal of Periodontal Research | 2007

Tumor necrosis factor-α stimulates gingival epithelial cells to release high mobility-group box 1

Yoko Morimoto; Ko-ichi Kawahara; Salunya Tancharoen; Kiyoshi Kikuchi; Takashi Matsuyama; Teruto Hashiguchi; Yuichi Izumi; Ikuro Maruyama

Collaboration


Dive into the Yoko Morimoto'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