2024/02/02 更新

写真a

ワン デンリ
王 登莉
WANG Dengli
所属
医歯薬学域 助教
職名
助教
連絡先
メールアドレス
外部リンク

学位

  • 医学博士 ( 2017年9月   岡山大学 )

研究キーワード

  • 脳出血

  • 炎症

  • 抗HMGB1抗体

研究分野

  • ライフサイエンス / 薬理学  / 脳出血 HMGB1 抗体

  • ライフサイエンス / 薬理学  / リボヌクレオタンパク質、創薬

学歴

  • 岡山大学   Graduate School of Medicine , Dentistry and Pharmaceutical Sciences  

    2014年10月 - 2017年9月

      詳細を見る

    国名: 日本国

    researchmap

  • 吉林大学 (中国)    

    2010年8月 - 2013年12月

      詳細を見る

    国名: 中華人民共和国

    researchmap

  • 長春中医薬大学 (中国)    

    2006年8月 - 2010年8月

      詳細を見る

経歴

  • 岡山大学   学術研究院医歯薬学域 薬理学   助教

    2021年4月 - 現在

      詳細を見る

  • 岡山大学   大学院医歯薬学総合研究科 薬理学   助教

    2017年10月 - 2021年3月

      詳細を見る

所属学協会

  • 日本癌学会

    2022年10月 - 現在

      詳細を見る

  • 日本神経科学学会

    2018年3月 - 現在

      詳細を見る

  • 北米神経科学学会

    2017年8月 - 現在

      詳細を見る

  • 日本薬理学会

    2014年8月 - 現在

      詳細を見る

 

論文

  • Perineural Treatment with High Mobility Group Box-1 Monoclonal Antibody Prevents Initiation of Pain-Like Behaviors in Female Mice with Trigeminal Neuropathy

    Simeng Ma, Yoki Nakamura, Takahiro Kochi, Suzuna Uemoto, Kazue Hisaoka-Nakashima, Dengli Wang, Keyue Liu, Hidenori Wake, Masahiro Nishibori, Norimitsu Morioka

    Biological and Pharmaceutical Bulletin   47 ( 1 )   221 - 226   2024年1月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:Pharmaceutical Society of Japan  

    DOI: 10.1248/bpb.b23-00729

    researchmap

  • Protective effects of an anti-4-HNE monoclonal antibody against liver injury and lethality of endotoxemia in mice

    Handong Qiao, Yuta Morioka, Dengli Wang, Keyue Liu, Shangze Gao, Hidenori Wake, Daiki Ousaka, Kiyoshi Teshigawara, Shuji Mori, Masahiro Nishibori

    European Journal of Pharmacology   950   175702 - 175702   2023年7月

     詳細を見る

    掲載種別:研究論文(学術雑誌)   出版者・発行元:Elsevier BV  

    DOI: 10.1016/j.ejphar.2023.175702

    researchmap

  • Histamine induced high mobility group box-1 release from vascular endothelial cells through H-1 receptor

    Shangze Gao, Keyue Liu, Wenhan Ku, Dengli Wang, Hidenori Wake, Handong Qiao, Kiyoshi Teshigawara, Masahiro Nishibori

    FRONTIERS IN IMMUNOLOGY   13   2022年10月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:FRONTIERS MEDIA SA  

    BackgroundSystemic allergic reaction is characterized by vasodilation and vascular leakage, which causes a rapid, precipitous and sustained decrease in arterial blood pressure with a concomitant decrease of cardiac output. Histamine is a major mediator released by mast cells in allergic inflammation and response. It causes a cascade of inflammation and strongly increases vascular permeability within minutes through its four G-protein-coupled receptors (GPCRs) on endothelial cells. High mobility group box-1 (HMGB1), a nonhistone chromatin-binding nuclear protein, can be actively secreted into the extracellular space by endothelial cells. HMGB1 has been reported to exert pro-inflammatory effects on endothelial cells and to increase vascular endothelial permeability. However, the relationship between histamine and HMGB1-mediated signaling in vascular endothelial cells and the role of HMGB1 in anaphylactic-induced hypotension have never been studied. Methods and resultsEA.hy 926 cells were treated with different concentrations of histamine for the indicated periods. The results showed that histamine induced HMGB1 translocation and release from the endothelial cells in a concentration- and time-dependent manner. These effects of histamine were concentration-dependently inhibited by d-chlorpheniramine, a specific H-1 receptor antagonist, but not by H-2 or H-3/4 receptor antagonists. Moreover, an H-1-specific agonist, 2-pyridylethylamine, mimicked the effects of histamine, whereas an H-2-receptor agonist, 4-methylhistamine, did not. Adrenaline and noradrenaline, which are commonly used in the clinical treatment of anaphylactic shock, also inhibited the histamine-induced HMGB1 translocation in endothelial cells. We therefore established a rat model of allergic shock by i.v. injection of compound 48/80, a potent histamine-releasing agent. The plasma HMGB1 levels in compound 48/80-injected rats were higher than those in controls. Moreover, the treatment with anti-HMGB1 antibody successfully facilitated the recovery from compound 48/80-induced hypotension. ConclusionHistamine induces HMGB1 release from vascular endothelial cells solely through H-1 receptor stimulation. Anti-HMGB1 therapy may provide a novel treatment for life-threatening systemic anaphylaxis.

    DOI: 10.3389/fimmu.2022.930683

    Web of Science

    researchmap

  • Treatment of Marmoset Intracerebral Hemorrhage with Humanized Anti-HMGB1 mAb

    Dengli Wang, Daiki Ousaka, Handong Qiao, Ziyi Wang, Kun Zhao, Shangze Gao, Keyue Liu, Kiyoshi Teshigawara, Kenzo Takada, Masahiro Nishibori

    CELLS   11 ( 19 )   2022年10月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:MDPI  

    Intracerebral hemorrhage (ICH) is recognized as a severe clinical problem lacking effective treatment. High mobility group box-1 (HMGB1) exhibits inflammatory cytokine-like activity once released into the extracellular space from the nuclei. We previously demonstrated that intravenous injection of rat anti-HMGB1 monoclonal antibody (mAb) remarkably ameliorated brain injury in a rat ICH model. Therefore, we developed a humanized anti-HMGB1 mAb (OKY001) for clinical use. The present study examined whether and how the humanized anti-HMGB1 mAb ameliorates ICH injury in common marmosets. The results show that administration of humanized anti-HMGB1 mAb inhibited HMGB1 release from the brain into plasma, in association with a decrease of 4-hydroxynonenal (4-HNE) accumulation and a decrease in cerebral iron deposition. In addition, humanized anti-HMGB1 mAb treatment resulted in a reduction in brain injury volume at 12 d after ICH induction. Our in vitro experiment showed that recombinant HMGB1 inhibited hemoglobin uptake by macrophages through CD163 in the presence of haptoglobin, suggesting that the release of excess HMGB1 from the brain may induce a delay in hemoglobin scavenging, thereby allowing the toxic effects of hemoglobin, heme, and Fe2+ to persist. Finally, humanized anti-HMGB1 mAb reduced body weight loss and improved behavioral performance after ICH. Taken together, these results suggest that intravenous injection of humanized anti-HMGB1 mAb has potential as a novel therapeutic strategy for ICH.

    DOI: 10.3390/cells11192970

    Web of Science

    researchmap

  • High-mobility group box 1-mediated hippocampal microglial activation induces cognitive impairment in mice with neuropathic pain

    Kazue Hisaoka-Nakashima, Kazuto Ohata, Natsuki Yoshimoto, Shintarou Tokuda, Nanako Yoshii, Yoki Nakamura, Dengli Wang, Keyue Liu, Hidenori Wake, Takayuki Yoshida, Yukio Ago, Kouichi Hashimoto, Masahiro Nishibori, Norimitsu Morioka

    Experimental Neurology   355   114146 - 114146   2022年9月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:Elsevier BV  

    Clinical evidence indicates that cognitive impairment is a common comorbidity of chronic pain, including neuropathic pain, but the mechanism underlying cognitive impairment remains unclear. Neuroinflammation plays a critical role in the development of both neuropathic pain and cognitive impairment. High-mobility group box 1 (HMGB1) is a proinflammatory molecule and could be involved in neuroinflammation-mediated cognitive impairment in the neuropathic pain state. Hippocampal microglial activation in mice has been associated with cognitive impairment. Thus, the current study examined a potential role of HMGB1 and microglial activation in cognitive impairment in mice with neuropathic pain due to a partial sciatic nerve ligation (PSNL). Mice developed cognitive impairment over two weeks, but not one week, after nerve injury. Nerve-injured mice demonstrated decreased nuclear fraction HMGB1, suggesting increased extracellular release of HMGB1. Furthermore, two weeks after PSNL, significant microglia activation was observed in hippocampus. Inhibition of microglial activation with minocycline, local hippocampal microglia depletion with clodronate liposome, or blockade of HMGB1 with either glycyrrhizic acid (GZA) or anti-HMGB1 antibody in PSNL mice reduced hippocampal microglia activation and ameliorated cognitive impairment. Other changes in the hippocampus of PSNL mice potentially related to cognitive impairment, including decreased hippocampal neuron dendrite length and spine densities and decreased alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptor (AMPAR) subunits, were prevented with anti-HMGB1 antibody treatment. The current findings suggest that neuro-inflammation involves a number of cellular-level changes and microglial activation. Blocking neuroinflammation, particularly through blocking HMGB1 could be a novel approach to reducing co-morbidities such as cognitive impairment associated with neuropathic pain.

    DOI: 10.1016/j.expneurol.2022.114146

    Web of Science

    researchmap

  • Development of hepatic impairment aggravates chemotherapy-induced peripheral neuropathy following oxaliplatin treatment: Evidence from clinical and preclinical studies.

    Tomoyoshi Miyamoto, Risa Domoto, Fumiko Sekiguchi, Riki Kamaguchi, Rika Nishimura, Misato Matsuno, Maho Tsubota, Masanori Fujitani, Shigekatsu Hatanaka, Yuichi Koizumi, Dengli Wang, Masahiro Nishibori, Atsufumi Kawabata

    Journal of pharmacological sciences   148 ( 3 )   315 - 325   2022年3月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:JAPANESE PHARMACOLOGICAL SOC  

    Oxaliplatin often induces peripheral neuropathy, a dose-limiting adverse reaction, and in rare cases leads to sinusoidal obstruction syndrome. We thus conducted a retrospective cohort study to examine the relationship between oxaliplatin-induced peripheral neuropathy (OIPN) and hepatic impairment, and then perform a fundamental study to analyze the underlying mechanisms. Analysis of medical records in cancer patients treated with oxaliplatin indicated that laboratory test parameters of hepatic impairment including AST, ALT and APRI (AST to platelet ratio index) moderately increased during oxaliplatin treatment, which was positively correlated with the severity of OIPN (grades 1-4), and associated with later incidence of survivors with OIPN grades ≥2. In mice, hepatic injury induced by CCl4 or ethanol accelerated OIPN in mice, an effect prevented by inactivation of high mobility group box 1 (HMGB1), known to participate in OIPN, by the neutralizing antibody or thrombomodulin alfa capable of promoting its thrombin-dependent degradation. Oxaliplatin also aggravated the hepatic injury in mice. CCl4 released HMGB1 from cultured hepatic parenchymal cells, and oxaliplatin at clinically achievable concentrations released HMGB1 from hepatic parenchymal and non-parenchymal cells. Our clinical and preclinical data suggest that the development of mild hepatic impairment during oxaliplatin treatment is associated with later aggravation of OIPN.

    DOI: 10.1016/j.jphs.2022.01.006

    Web of Science

    PubMed

    researchmap

  • Targeting necroptosis in muscle fibers ameliorates inflammatory myopathies

    Mari Kamiya, Fumitaka Mizoguchi, Kimito Kawahata, Dengli Wang, Masahiro Nishibori, Jessica Day, Cynthia Louis, Ian P. Wicks, Hitoshi Kohsaka, Shinsuke Yasuda

    NATURE COMMUNICATIONS   13 ( 1 )   2022年1月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:NATURE PORTFOLIO  

    Muscle cell death in polymyositis is induced by CD8(+) cytotoxic T lymphocytes. We hypothesized that the injured muscle fibers release pro-inflammatory molecules, which would further accelerate CD8(+) cytotoxic T lymphocytes-induced muscle injury, and inhibition of the cell death of muscle fibers could be a novel therapeutic strategy to suppress both muscle injury and inflammation in polymyositis. Here, we show that the pattern of cell death of muscle fibers in polymyositis is FAS ligand-dependent necroptosis, while that of satellite cells and myoblasts is perforin 1/granzyme B-dependent apoptosis, using human muscle biopsy specimens of polymyositis patients and models of polymyositis in vitro and in vivo. Inhibition of necroptosis suppresses not only CD8(+) cytotoxic T lymphocytes-induced cell death of myotubes but also the release of inflammatory molecules including HMGB1. Treatment with a necroptosis inhibitor or anti-HMGB1 antibodies ameliorates myositis-induced muscle weakness as well as muscle cell death and inflammation in the muscles. Thus, targeting necroptosis in muscle cells is a promising strategy for treating polymyositis providing an alternative to current therapies directed at leukocytes.

    DOI: 10.1038/s41467-021-27875-4

    Web of Science

    researchmap

  • Central high mobility group box-1 induces mechanical hypersensitivity with spinal microglial activation in a mouse model of hemi-Parkinson’s disease

    Fumiaki Sato, Yoki Nakamura, Simeng Ma, Takahiro Kochi, Kazue Hisaoka-Nakashima, Dengli Wang, Keyue Liu, Hidenori Wake, Masahiro Nishibori, Norimitsu Morioka

    Biomedicine & Pharmacotherapy   145   112479 - 112479   2022年1月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:Elsevier BV  

    Parkinson's disease (PD) patients often complain of pain, but this problem has been neglected and is poorly understood. High mobility group box-1 (HMGB1), an alarmin/damage-associated molecular patterns protein, is increased in the cerebrospinal fluid in PD patients. However, little is known of the relationship between HMGB1 and pain associated with PD. Here, we investigated the role of central HMGB1 in the regulation of nociceptive hypersensitivity in a mouse model of PD. Male ddY mice were microinjected unilaterally with 6-hydroxydop-amine (6OHDA) into the striatum. These hemi-PD mice were treated with anti-HMGB1 neutralizing antibody (nAb; 10 mu g in 10 mu L) by intranasal (i.n.) administration. The mechanical hypersensitivity of the hind paws was evaluated with the von Frey test. Spinal microglial activity was analyzed by immunostaining for ionized calcium-binding adapter molecule 1. The 6OHDA-administered mice displayed unilateral loss of dopamine neurons in the substantia nigra and mechanical hypersensitivity in both hind paws. Moreover, spinal microglia were activated in these hemi-PD mice. Twenty-eight days after the 6OHDA injections, repeated i.n., but not systemic, treatment with anti-HMGB1 nAb inhibited the bilateral mechanical hypersensitivity and spinal microglial activation. However, the anti-HMGB1 nAb did not ameliorate the dopamine neuron loss. Moreover, intracerebroventricular injection with recombinant HMGB1 induced mechanical hypersensitivity. These findings indicate that HMGB1 is involved in the maintenance of nociceptive symptoms in hemi-PD mice via spinal microglial activation. Therefore, central HMGB1 may have potential as a therapeutic target for pain associated with PD.

    DOI: 10.1016/j.biopha.2021.112479

    Web of Science

    researchmap

  • Role of neuron-derived ATP in paclitaxel-induced HMGB1 release from macrophages and peripheral neuropathy.

    Risa Domoto, Fumiko Sekiguchi, Riki Kamaguchi, Maiko Iemura, Hiroki Yamanishi, Maho Tsubota, Dengli Wang, Masahiro Nishibori, Atsufumi Kawabata

    Journal of pharmacological sciences   148 ( 1 )   156 - 161   2022年1月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:JAPANESE PHARMACOLOGICAL SOC  

    We examined the role of ATP and high mobility group box 1 (HMGB1) in paclitaxel-induced peripheral neuropathy (PIPN). PIPN in mice was prevented by HMGB1 neutralization, macrophage depletion, and P2X7 or P2X4 blockade. Paclitaxel and ATP synergistically released HMGB1 from macrophage-like RAW264.7 cells, but not neuron-like NG108-15 cells. The paclitaxel-induced HMGB1 release from RAW264.7 cells was accelerated by co-culture with NG108-15 cells in a manner dependent on P2X7 or P2X4. Paclitaxel released ATP from NG108-15 cells, but not RAW264.7 cells. Thus, PIPN is considered to involve acceleration of HMGB1 release from macrophages through P2X7 and P2X4 activation by neuron-derived ATP.

    DOI: 10.1016/j.jphs.2021.11.003

    Web of Science

    PubMed

    researchmap

  • 溶血に伴うDAMPs処理をターゲットにした敗血症(COVID-19含む)に対する治療法の開発

    逢坂 大樹, 王 登莉, 西堀 正洋

    日本薬理学会年会要旨集   95   1-S03-1   2022年

     詳細を見る

    記述言語:日本語   出版者・発行元:公益社団法人 日本薬理学会  

    Sepsis is one of the leading cause of death worldwide. Recently, several studies suggested that free-hemoglobin and heme derived from hemolysis are important factors which may be associated with severity of septic patients including COVID-19. In other words, hemolysis-derived products enhance the inflammatory responses as damage-associated molecular patterns (DAMPs) in both intravascular and extravascular space. In addition, hemoglobin has vasoconstrictive activity by depleting nitric oxide, whereas heme or Fe2+ produce reactive oxygen species (ROS) through Fenton reaction leading to tissue injury. At present, we have no therapeutic options against sepsis-related hemolysis in clinical settings, however, there are might be two therapeutic strategies in this regard. One is supplemental therapy of depleted scavenging proteins such as haptoglobin and hemopexin, the other is activation of the internal scavenging system including macrophage-CD163 pathway. These novel targets against sepsis are also critical for the next pandemic. We will show you our recent data aiming at anti-hemolytic therapy.

    DOI: 10.1254/jpssuppl.95.0_1-s03-1

    researchmap

  • Combinatrial treatment of anti-High Mobility Group Box-1 monoclonal antibody and epothilone B improves functional recovery after spinal cord contusion injury. 国際誌

    Yicheng Zhu, Naohiro Uezono, Tetsuro Yasui, Masahide Nakajo, Tatsuya Nagai, Dengli Wang, Masahiro Nishibori, Kinichi Nakashima

    Neuroscience research   172   13 - 25   2021年11月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:ELSEVIER IRELAND LTD  

    Spinal cord injury (SCI) causes motor and sensory deficits and is currently considered an incurable disease. We have previously reported that administration of anti-High Mobility Group Box-1 monoclonal antibody (anti-HMGB1 mAb) preserved lesion area and improved locomotion recovery in mouse model of SCI. In order to further enhance the recovery, we here examined combinatorial treatment of anti-HMGB1 mAb and epothilone B (Epo B), which has been reported to promote axon regeneration. This combinatorial treatment significantly increased hindlimb movement compared with anti-HMGB1 mAb alone, although Epo B alone failed to increase functional recovery. These results are in agreement with that anti-HMGB1 mAb alone was able to decrease the lesion area spreading and increase the surviving neuron numbers around the lesion, whereas Epo B facilitated axon outgrowth only in combination with anti-HMGB1 mAb, suggesting that anti-HMGB1 mAb-dependent tissue preservation is necessary for Epo B to exhibit its therapeutic effect. Taken together, the combinatorial treatment can be considered as a novel and clinically applicable strategy for SCI.

    DOI: 10.1016/j.neures.2021.04.002

    Web of Science

    PubMed

    researchmap

  • Neuroprotective Effects of Anti-high Mobility Group Box-1 Monoclonal Antibody Against Methamphetamine-Induced Dopaminergic Neurotoxicity. 国際誌

    Kaori Masai, Keita Kuroda, Nami Isooka, Ryo Kikuoka, Shinki Murakami, Sunao Kamimai, Dengli Wang, Keyue Liu, Ikuko Miyazaki, Masahiro Nishibori, Masato Asanuma

    Neurotoxicity research   39 ( 5 )   1511 - 1523   2021年10月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:SPRINGER  

    High mobility group box-1 (HMGB1) is a ubiquitous non-histone nuclear protein that plays a key role as a transcriptional activator, with its extracellular release provoking inflammation. Inflammatory responses are essential in methamphetamine (METH)-induced acute dopaminergic neurotoxicity. In the present study, we examined the effects of neutralizing anti-HMGB1 monoclonal antibody (mAb) on METH-induced dopaminergic neurotoxicity in mice. BALB/c mice received a single intravenous administration of anti-HMGB1 mAb prior to intraperitoneal injections of METH (4 mg/kg × 2, at 2-h intervals). METH injections induced hyperthermia, an increase in plasma HMGB1 concentration, degeneration of dopaminergic nerve terminals, accumulation of microglia, and extracellular release of neuronal HMGB1 in the striatum. These METH-induced changes were significantly inhibited by intravenous administration of anti-HMGB1 mAb. In contrast, blood-brain barrier disruption occurred by METH injections was not suppressed. Our findings demonstrated the neuroprotective effects of anti-HMGB1 mAb against METH-induced dopaminergic neurotoxicity, suggesting that HMGB1 could play an initially important role in METH toxicity.

    DOI: 10.1007/s12640-021-00402-5

    Web of Science

    PubMed

    researchmap

  • Caspase-Dependent HMGB1 Release from Macrophages Participates in Peripheral Neuropathy Caused by Bortezomib, a Proteasome-Inhibiting Chemotherapeutic Agent, in Mice. 国際誌

    Maho Tsubota, Takaya Miyazaki, Yuya Ikeda, Yusuke Hayashi, Yui Aokiba, Shiori Tomita, Fumiko Sekiguchi, Dengli Wang, Masahiro Nishibori, Atsufumi Kawabata

    Cells   10 ( 10 )   2021年9月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:MDPI  

    Given the role of macrophage-derived high mobility group box 1 (HMGB1) in chemotherapy-induced peripheral neuropathy (CIPN) caused by paclitaxel, we analyzed the role of HMGB1 and macrophages in the CIPN caused by bortezomib, a proteasome-inhibiting chemotherapeutic agent used for the treatment of multiple myeloma. Repeated administration of bortezomib caused CIPN accompanied by early-stage macrophage accumulation in the dorsal root ganglion. This CIPN was prevented by an anti-HMGB1-neutralizing antibody, thrombomodulin alfa capable of accelerating thrombin-dependent degradation of HMGB1, antagonists of the receptor for advanced glycation end-products (RAGE) and C-X-C motif chemokine receptor 4 (CXCR4), known as HMGB1-targeted membrane receptors, or macrophage depletion with liposomal clodronate, as reported in a CIPN model caused by paclitaxel. In macrophage-like RAW264.7 cells, bortezomib as well as MG132, a well-known proteasome inhibitor, caused HMGB1 release, an effect inhibited by caspase inhibitors but not inhibitors of NF-κB and p38 MAP kinase, known to mediate paclitaxel-induced HMGB1 release from macrophages. Bortezomib increased cleaved products of caspase-8 and caused nuclear fragmentation or condensation in macrophages. Repeated treatment with the caspase inhibitor prevented CIPN caused by bortezomib in mice. Our findings suggest that bortezomib causes caspase-dependent release of HMGB1 from macrophages, leading to the development of CIPN via activation of RAGE and CXCR4.

    DOI: 10.3390/cells10102550

    Web of Science

    PubMed

    researchmap

  • Pretreatment with High Mobility Group Box-1 Monoclonal Antibody Prevents the Onset of Trigeminal Neuropathy in Mice with a Distal Infraorbital Nerve Chronic Constriction Injury. 国際誌

    Takahiro Kochi, Yoki Nakamura, Simeng Ma, Kazue Hisaoka-Nakashima, Dengli Wang, Keyue Liu, Hidenori Wake, Masahiro Nishibori, Masahiro Irifune, Norimitsu Morioka

    Molecules (Basel, Switzerland)   26 ( 7 )   2021年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:MDPI  

    Persistent pain following orofacial surgery is not uncommon. High mobility group box 1 (HMGB1), an alarmin, is released by peripheral immune cells following nerve injury and could be related to pain associated with trigeminal nerve injury. Distal infraorbital nerve chronic constriction injury (dIoN-CCI) evokes pain-related behaviors including increased facial grooming and hyper-responsiveness to acetone (cutaneous cooling) after dIoN-CCI surgery in mice. In addition, dIoN-CCI mice developed conditioned place preference to mirogabalin, suggesting increased neuropathic pain-related aversion. Treatment of the infraorbital nerve with neutralizing antibody HMGB1 (anti-HMGB1 nAb) before dIoN-CCI prevented both facial grooming and hyper-responsiveness to cooling. Pretreatment with anti-HMGB1 nAb also blocked immune cell activation associated with trigeminal nerve injury including the accumulation of macrophage around the injured IoN and increased microglia activation in the ipsilateral spinal trigeminal nucleus caudalis. The current findings demonstrated that blocking of HMGB1 prior to nerve injury prevents the onset of pain-related behaviors, possibly through blocking the activation of immune cells associated with the nerve injury, both within the CNS and on peripheral nerves. The current findings further suggest that blocking HMGB1 before tissue injury could be a novel strategy to prevent the induction of chronic pain following orofacial surgeries.

    DOI: 10.3390/molecules26072035

    Web of Science

    PubMed

    researchmap

  • Perineural high-mobility group box 1 induces mechanical hypersensitivity through activation of spinal microglia: Involvement of glutamate-NMDA receptor dependent mechanism in spinal dorsal horn. 国際誌

    Yoki Nakamura, Ayako Fukuta, Keita Miyashita, Fang Fang Zhang, Dengli Wang, Keyue Liu, Hidenori Wake, Kazue Hisaoka-Nakashima, Masahiro Nishibori, Norimitsu Morioka

    Biochemical pharmacology   186   114496 - 114496   2021年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:PERGAMON-ELSEVIER SCIENCE LTD  

    High mobility box 1 (HMGB1), a damage-associated molecular pattern, has crucial roles in induction of neuropathic pain. Upregulation of HMGB1 around the injured sciatic nerve contributes to mechanical hypersensitivity following partial sciatic nerve ligation (PSNL) of mice. However, central mechanisms mediating perineural HMGB1-induced nociceptive hypersensitivity, especially within the spinal dorsal horn, have not been determined. The current study shows that perineural treatment of naïve mice with recombinant HMGB1, which mimics increased HMGB1 around the injured sciatic nerve of PSNL mice, significantly induced activation of microglia, but not astrocytes, in the spinal dorsal horn. Intraperitoneal injection of minocycline, a microglial inhibitor, ameliorated perineural rHMGB1-induced mechanical hypersensitivity. In addition, blockade of spinal N-methyl-D-aspartate (NMDA) receptors significantly prevented perineural rHMGB1-induced mechanical hypersensitivity and microglial activation. In contrast, non-NMDA receptors, neurokinin 1 receptor, colony-stimulating factor 1 receptor and P2Y12 receptor were not involved in perineural rHMGB1-induced mechanical hypersensitivity. Furthermore, repeated perineural treatment with an anti-HMGB1 antibody blocked activation of spinal microglia in PSNL mice. Collectively, the current findings demonstrate that increased HMGB1 around injured sciatic nerve might induce nociceptive hypersensitivity through activation of spinal microglia. Thus, HMGB1-dependent mechanisms between the injured sciatic nerve and spinal dorsal horn could be crucial in induction of neuropathic pain.

    DOI: 10.1016/j.bcp.2021.114496

    Web of Science

    PubMed

    researchmap

  • Histidine-Rich Glycoprotein Stimulates Human Neutrophil Phagocytosis and Prolongs Survival through CLEC1A. 国際誌

    Yohei Takahashi, Hidenori Wake, Masakiyo Sakaguchi, Yukinori Yoshii, Kiyoshi Teshigawara, Dengli Wang, Masahiro Nishibori

    Journal of immunology (Baltimore, Md. : 1950)   206 ( 4 )   737 - 750   2021年2月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER ASSOC IMMUNOLOGISTS  

    Histidine-rich glycoprotein (HRG) is a multifunctional plasma protein and maintains the homeostasis of blood cells and vascular endothelial cells. In the current study, we demonstrate that HRG and recombinant HRG concentration dependently induced the phagocytic activity of isolated human neutrophils against fluorescence-labeled Escherichia coli and Staphylococcus aureus through the stimulation of CLEC1A receptors, maintaining their spherical round shape. The phagocytosis-inducing effects of HRG were inhibited by a specific anti-HRG Ab and enhanced by opsonization of bacteria with diluted serum. HRG and C5a prolonged the survival time of isolated human neutrophils, in association with a reduction in the spontaneous production of extracellular ROS. In contrast, HRG maintained the responsiveness of neutrophils to TNF-α, zymosan, and E. coli with regard to reactive oxygen species production. The blocking Ab for CLEC1A and recombinant CLEC1A-Fc fusion protein significantly inhibited the HRG-induced neutrophil rounding, phagocytic activity, and prolongation of survival time, suggesting the involvement of the CLEC1A receptor in the action of HRG on human neutrophils. These results as a whole indicated that HRG facilitated the clearance of E. coli and S. aureus by maintaining the neutrophil morphology and phagocytosis, contributing to the antiseptic effects of HRG in vivo.

    DOI: 10.4049/jimmunol.2000817

    Web of Science

    PubMed

    researchmap

  • 脊髄虚血再灌流障害に対する抗HMGB-1抗体の有効性の検討

    村岡 玄哉, 劉 克約, 喬 寒棟, 逢坂 大樹, 王 登莉, 藤井 泰宏, 笠原 真悟, 西堀 正洋

    日本薬理学会年会要旨集   94   3-Y-F2-1   2021年

     詳細を見る

    記述言語:日本語   出版者・発行元:公益社団法人 日本薬理学会  

    Introduction

    Spinal cord ischemia and reperfusion (I/R) injury is one of the most devastating complications after thoracic and thoracoabdominal aortic aneurysm surgery. Several studies showed that levels of HMGB1 in serum were increased in patients who suffered from spinal cord I/R injury. These data strongly suggest that HMGB1 may play a crucial role in spinal cord I/R injury.

    Materials and Methods

    Male New Zealand white rabbits were used for the experiments. The abdominal aorta at the level of the left renal artery was exposed, and, after heparinization, occluded for 11 minutes. The rabbits were intravenously administered an anti-HMGB1 mAb (#10-22, 2mg/kg) or class-matched control IgG (anti-keyhole limpet hemocyanin mAb) twice immediately and 6h after reperfusion. The neurological findings were assessed at 6, 24 and 48 hours after reperfusion. At 48hr after reperfusion, the rabbits were sacrificed to obtain the specimen of spinal cord and blood samples.

    Results

    Administration of anti-HMGB1 mAb ameliorated the intensity of spinal cord infarction and preserved the number of motor neuron cells, in association with decreased activated microglia and astrocyte. Consequently, anti-HMGB1 mAb significantly improved the neurological outcomes after spinal I/R injury in rabbits.  These results strongly indicate that HMGB1 plays a critical role in the development of spinal cord I/R induced secondary injury through the amplification of inflammatory response.

    Conclusion

    Intravenous injection of neutralizing anti-HMGB1 mAb has potential as a novel therapeutic strategy for spinal cord I/R injury.

    DOI: 10.1254/jpssuppl.94.0_3-y-f2-1

    CiNii Article

    researchmap

  • High Mobility Group Box-1 and Blood-Brain Barrier Disruption. 国際誌

    Masahiro Nishibori, Dengli Wang, Daiki Ousaka, Hidenori Wake

    Cells   9 ( 12 )   2020年12月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:MDPI  

    Increasing evidence suggests that inflammatory responses are involved in the progression of brain injuries induced by a diverse range of insults, including ischemia, hemorrhage, trauma, epilepsy, and degenerative diseases. During the processes of inflammation, disruption of the blood-brain barrier (BBB) may play a critical role in the enhancement of inflammatory responses and may initiate brain damage because the BBB constitutes an interface between the brain parenchyma and the bloodstream containing blood cells and plasma. The BBB has a distinct structure compared with those in peripheral tissues: it is composed of vascular endothelial cells with tight junctions, numerous pericytes surrounding endothelial cells, astrocytic endfeet, and a basement membrane structure. Under physiological conditions, the BBB should function as an important element in the neurovascular unit (NVU). High mobility group box-1 (HMGB1), a nonhistone nuclear protein, is ubiquitously expressed in almost all kinds of cells. HMGB1 plays important roles in the maintenance of chromatin structure, the regulation of transcription activity, and DNA repair in nuclei. On the other hand, HMGB1 is considered to be a representative damage-associated molecular pattern (DAMP) because it is translocated and released extracellularly from different types of brain cells, including neurons and glia, contributing to the pathophysiology of many diseases in the central nervous system (CNS). The regulation of HMGB1 release or the neutralization of extracellular HMGB1 produces beneficial effects on brain injuries induced by ischemia, hemorrhage, trauma, epilepsy, and Alzheimer's amyloidpathy in animal models and is associated with improvement of the neurological symptoms. In the present review, we focus on the dynamics of HMGB1 translocation in different disease conditions in the CNS and discuss the functional roles of extracellular HMGB1 in BBB disruption and brain inflammation. There might be common as well as distinct inflammatory processes for each CNS disease. This review will provide novel insights toward an improved understanding of a common pathophysiological process of CNS diseases, namely, BBB disruption mediated by HMGB1. It is proposed that HMGB1 might be an excellent target for the treatment of CNS diseases with BBB disruption.

    DOI: 10.3390/cells9122650

    Web of Science

    PubMed

    researchmap

  • An Evaluation of the Activity of Histidine-Rich Glycoprotein on Differentiated Neutrophil-Like Cells from Human Cell Lines. 国際誌

    Yukinori Yoshii, Hidenori Wake, Yoshito Nishimura, Kiyoshi Teshigawara, Dengli Wang, Masahiro Nishibori

    The Journal of pharmacology and experimental therapeutics   375 ( 3 )   406 - 413   2020年12月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:AMER SOC PHARMACOLOGY EXPERIMENTAL THERAPEUTICS  

    Histidine-rich glycoprotein (HRG) treatment ameliorated the survival rate of septic mice by suppressing excess immunothrombus formation. Although such findings suggested that HRG may be one of the most useful drugs for sepsis, obtaining a stable experimental system to standardize the HRG drug product is difficult to achieve using neutrophils isolated from volunteers. This is due to the short survival time and individual differences of human neutrophils. In the present study, we determined whether the differentiated neutrophil-like cell lines exhibited similar responses to HRG compared with human purified neutrophils. All-trans retinoic acid (ATRA) was employed to induce the differentiation of the human myeloid leukemia cell lines HL-60 and NB-4. Thereafter, the cells were treated with Hank's balanced salt solution, human serum albumin, or HRG. The effects of HRG on these cells were evaluated according to cell shape, microcapillary passage, reactive oxygen species (ROS) production, neutrophil extracellular traps (NETs) formation, the expression of activated CD11b, and cell viability. HRG maintained the round shape of differentiated neutrophil-like cells, decreased the time required by cells to pass through the microcapillaries, and inhibited ROS production, NETs formation, and the expression of activated CD11b on the cell surface. Moreover, the cells could survive longer in the presence of HRG than the control. The ATRA-induced differentiated cell lines could be used as alternatives to neutrophils to investigate the effects of HRG on neutrophils. This method can thus be used as an essential standardization test in pharmaceutical development. SIGNIFICANCE STATEMENT: Human neutrophils exhibit varying responses to histidine-rich glycoprotein (HRG); however, all-trans retinoic acid-induced differentiated neutrophil-like cell lines can be used as reliably proxies to investigate the effects of HRG on neutrophils. Additionally, these cell lines can be employed in the development of therapies for the treatment of sepsis.

    DOI: 10.1124/jpet.120.000182

    Web of Science

    PubMed

    researchmap

  • Histidine-rich glycoprotein possesses antioxidant activity through self-oxidation and inhibition of hydroxyl radical production via chelating divalent metal ions in Fenton's reaction. 国際誌

    Hidenori Wake, Yohei Takahashi, Yukinori Yoshii, Shangze Gao, Shuji Mori, Dengli Wang, Kiyoshi Teshigawara, Masahiro Nishibori

    Free radical research   54 ( 8-9 )   649 - 661   2020年9月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:TAYLOR & FRANCIS LTD  

    Sepsis is caused by infections associated with life-threatening multiple organ failure (MOF). Septic MOF appears to be closely related to circulatory failure due to immunothrombosis. This process involves the production of reactive oxygen spices (ROS) in inflammatory sites. Therefore, the detoxification of the systemic excess ROS is important for the improvement of the process in septic pathogenesis. Histidine-rich glycoprotein (HRG), a plasma glycoprotein, ameliorates a septic condition through the suppression of both excess ROS production from neutrophils and immunothrombosis. Hydroxyl radical is known as the most important species among ROS in pathogenesis; however, the direct influence of HRG on hydroxyl radical formation and ROS activity is poorly understood. In this study, we showed that HRG, in a concentration-dependent manner, efficiently inhibited the production of hydroxyl radical induced by the Fenton's reaction through chelation of the divalent iron. HRG also exhibited antioxidant activity against peroxyl radical by oxidation of HRG itself as a substrate; however, it did not show superoxide dismutase and catalase-like activities. Additionally, HRG enhanced glutathione peroxidase, a well-known antioxidant enzyme, activity. These results suggest that HRG may play a unique role in suppression of the production of hydroxyl radicals and subsequent tissue damage at inflammatory sites. Marked reduction in plasma HRG in sepsis might lose such an important protective mechanism. Thus, the present study provides evidence that inhibition of ROS and ROS-production systems by HRG may contribute to antiseptic effects in vivo and that HRG could be potential therapy for ROS-related diseases.

    DOI: 10.1080/10715762.2020.1825703

    Web of Science

    PubMed

    researchmap

  • Histidine-Rich Glycoprotein Inhibits High-Mobility Group Box-1-Mediated Pathways in Vascular Endothelial Cells through CLEC-1A. 国際誌

    Shangze Gao, Hidenori Wake, Masakiyo Sakaguchi, Dengli Wang, Youhei Takahashi, Kiyoshi Teshigawara, Hui Zhong, Shuji Mori, Keyue Liu, Hideo Takahashi, Masahiro Nishibori

    iScience   23 ( 6 )   101180 - 101180   2020年6月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:CELL PRESS  

    High-mobility group box-1 (HMGB1) protein has been postulated to play a pathogenic role in severe sepsis. Histidine-rich glycoprotein (HRG), a 75 kDa plasma protein, was demonstrated to improve the survival rate of septic mice through the regulation of neutrophils and endothelium barrier function. As the relationship of HRG and HMGB1 remains poorly understood, we investigated the effects of HRG on HMGB1-mediated pathway in endothelial cells, focusing on the involvement of specific receptors for HRG. HRG potently inhibited the HMGB1 mobilization and effectively suppressed rHMGB1-induced inflammatory responses and expression of all three HMGB1 receptors in endothelial cells. Moreover, we first clarified that these protective effects of HRG on endothelial cells were mediated through C-type lectin domain family 1 member A (CLEC-1A) receptor. Thus, current study elucidates protective effects of HRG on vascular endothelial cells through inhibition of HMGB1-mediated pathways may contribute to the therapeutic effects of HRG on severe sepsis.

    DOI: 10.1016/j.isci.2020.101180

    Web of Science

    PubMed

    researchmap

  • Corticosterone Induces HMGB1 Release in Primary Cultured Rat Cortical Astrocytes: Involvement of Pannexin-1 and P2X7 Receptor-Dependent Mechanisms. 国際誌

    Kazue Hisaoka-Nakashima, Honami Azuma, Fumina Ishikawa, Yoki Nakamura, Dengli Wang, Keyue Liu, Hidenori Wake, Masahiro Nishibori, Yoshihiro Nakata, Norimitsu Morioka

    Cells   9 ( 5 )   2020年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:MDPI  

    A major risk factor for major depressive disorder (MDD) is stress. Stress leads to the release of high-mobility group box-1 (HMGB1), which in turn leads to neuroinflammation, a potential pathophysiological basis of MDD. The mechanism underlying stress-induced HMGB1 release is not known, but stress-associated glucocorticoids could be involved. To test this, rat primary cultured cortical astrocytes, the most abundant cell type in the central nervous system (CNS), were treated with corticosterone and HMGB1 release was assessed by Western blotting and ELISA. Significant HMGB1 was released with treatment with either corticosterone or dexamethasone, a synthetic glucocorticoid. HMGB1 translocated from the nucleus to the cytoplasm following corticosterone treatment. HMGB1 release was significantly attenuated with glucocorticoid receptor blocking. In addition, inhibition of pannexin-1, and P2X7 receptors led to a significant decrease in corticosterone-induced HMGB1 release. Taken together, corticosterone stimulates astrocytic glucocorticoid receptors and triggers cytoplasmic translocation and extracellular release of nuclear HMGB1 through a mechanism involving pannexin-1 and P2X7 receptors. Thus, under conditions of stress, glucocorticoids induce astrocytic HMGB1 release, leading to a neuroinflammatory state that could mediate neurological disorders such as MDD.

    DOI: 10.3390/cells9051068

    Web of Science

    PubMed

    researchmap

  • HMGB1 Translocation in Neurons after Ischemic Insult: Subcellular Localization in Mitochondria and Peroxisomes. 国際誌

    Dengli Wang, Keyue Liu, Yusuke Fukuyasu, Kiyoshi Teshigawara, Li Fu, Hidenori Wake, Aiji Ohtsuka, Masahiro Nishibori

    Cells   9 ( 3 )   2020年3月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:MDPI  

    High mobility group box-1 (HMGB1), a nonhistone chromatin DNA-binding protein, is released from neurons into the extracellular space under ischemic, hemorrhagic, and traumatic insults. However, the details of the time-dependent translocation of HMGB1 and the subcellular localization of HMGB1 through the release process in neurons remain unclear. In the present study, we examined the subcellular localization of HMGB1 during translocation of HMGB1 in the cytosolic compartment using a middle cerebral artery occlusion and reperfusion model in rats. Double immunofluorescence microscopy revealed that HMGB1 immunoreactivities were colocalized with MTCO1(mitochondrially encoded cytochrome c oxidase I), a marker of mitochondria, and catalase, a marker of peroxisomes, but not with Rab5/Rab7 (RAS-related GTP-binding protein), LC3A/B (microtubule-associated protein 1 light chain 3), KDEL (KDEL amino acid sequence), and LAMP1 (Lysosomal Associated Membrane Protein 1), which are endosome, phagosome, endoplasmic reticulum, and lysosome markers, respectively. Immunoelectron microscopy confirmed that immune-gold particles for HMGB1 were present inside the mitochondria and peroxisomes. Moreover, HMGB1 was found to be colocalized with Drp1 (Dynamin-related protein 1), which is involved in mitochondrial fission. These results revealed the specific subcellular localization of HMGB1 during its release process under ischemic conditions.

    DOI: 10.3390/cells9030643

    Web of Science

    PubMed

    researchmap

  • Histidine-rich glycoprotein ameliorates endothelial barrier dysfunction through regulation of NF-κB and MAPK signal pathway. 国際誌

    Shangze Gao, Hidenori Wake, Yuan Gao, Dengli Wang, Shuji Mori, Keyue Liu, Kiyoshi Teshigawara, Hideo Takahashi, Masahiro Nishibori

    British journal of pharmacology   176 ( 15 )   2808 - 2824   2019年8月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:WILEY  

    BACKGROUND AND PURPOSE: Microvascular barrier breakdown is a hallmark of sepsis that is associated with sepsis-induced multiorgan failure. Histidine-rich glycoprotein (HRG) is a 75-kDa plasma protein that was demonstrated to improve the survival of septic mice through regulation of cell shape, spontaneous ROS production in neutrophils, and adhesion of neutrophils to vascular endothelial cells. We investigated HRG's role in the LPS/TNF-α-induced barrier dysfunction of endothelial cells in vitro and in vivo and the possible mechanism, to clarify the definitive roles of HRG in sepsis. EXPERIMENTAL APPROACH: EA.hy 926 endothelial cells were pretreated with HRG or human serum albumin before stimulation with LPS/TNF-α. A variety of biochemical assays were applied to explore the underlying molecular mechanisms on how HRG protected the barrier function of vascular endothelium. KEY RESULTS: Immunostaining results showed that HRG maintains the endothelial monolayer integrity by inhibiting cytoskeleton reorganization, losses of VE-cadherin and β-catenin, focal adhesion kinase degradation, and cell detachment induced by LPS/TNF-α. HRG also inhibited the cytokine secretion from endothelial cells induced by LPS/TNF-α, which was associated with reduced NF-κB activation. Moreover, HRG effectively prevented the LPS/TNF-α-induced increase in capillary permeability in vitro and in vivo. Finally, Western blot results demonstrated that HRG prevented the phosphorylation of MAPK family and RhoA activation, which are involved mainly in the regulation of cytoskeleton reorganization and barrier permeability. CONCLUSIONS AND IMPLICATIONS: Taken together, our results demonstrate that HRG has protective effects on vascular barrier function in vitro and in vivo, which may be due to the inhibition of MAPK family and Rho activation.

    DOI: 10.1111/bph.14711

    Web of Science

    PubMed

    researchmap

  • Histidine-rich glycoprotein augments natural killer cell function by modulating PD-1 expression via CLEC-1B. 国際誌

    Yoshito Nishimura, Hidenori Wake, Kiyoshi Teshigawara, Dengli Wang, Masakiyo Sakaguchi, Fumio Otsuka, Masahiro Nishibori

    Pharmacology research & perspectives   7 ( 3 )   e00481   2019年6月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:JOHN WILEY & SONS LTD  

    Augmentation of natural killer (NK) cell cytotoxicity is one of the greatest challenges for cancer immunotherapy. Although histidine-rich glycoprotein (HRG), a 75-kDa glycoprotein with various immunomodulatory activities, reportedly elicits antitumor immunity, its effect on NK cell cytotoxicity is unclear. We assessed NK cell cytotoxicity against K562 cells. We also measured concentrations of cytokines and granzyme B in the cell supernatant. The proportion of CD56bright NK cells and NK cell surface PD-1 expression was assessed with flow cytometry. The neutralizing effects of anti-C-type lectin-like receptor (CLEC) 1B against HRG were also measured. NK cell morphological changes were analyzed via confocal microscopy. HRG significantly increased NK cell cytotoxicity against K562 cell lines. HRG also increased the release of granzyme B and the proportion of CD56bright NK cells. Further, HRG was able to decrease NK cell surface PD-1 expression. The effects of HRG on NK cells were reversed with anti-CLEC-1B antibodies. Additionally, we confirmed NK cell nuclear morphology and F-actin distribution, which are involved in the regulation of cytotoxic granule secretion. Because both PD-1 and CLEC-1B are associated with prognosis during malignancy, HRG incorporates these molecules to exert the antitumor immunity role. These facts indicate the potential of HRG to be a new target for cancer immunotherapy.

    DOI: 10.1002/prp2.481

    Web of Science

    PubMed

    researchmap

  • The specific localization of advanced glycation end-products (AGEs) in rat pancreatic islets.

    Yuta Morioka, Kiyoshi Teshigawara, Yasuko Tomono, Dengli Wang, Yasuhisa Izushi, Hidenori Wake, Keyue Liu, Hideo Kohka Takahashi, Shuji Mori, Masahiro Nishibori

    Journal of pharmacological sciences   134 ( 4 )   218 - 224   2017年8月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:JAPANESE PHARMACOLOGICAL SOC  

    Advanced glycation end-products (AGEs) are produced by non-enzymatic glycation between protein and reducing sugar such as glucose. Although glyceraldehyde-derived AGEs (Glycer-AGEs), one of the AGEs subspecies, have been reported to be involved in the pathogenesis of various age-relating diseases such as diabetes mellitus or arteriosclerosis, little is known about the pathological and physiological mechanism of AGEs in vivo. In present study, we produced 4 kinds of polyclonal antibodies against AGEs subspecies and investigated the localization of AGEs-modified proteins in rat peripheral tissues, making use of these antibodies. We found that Glycer-AGEs and methylglyoxal-derived AGEs (MGO-AGEs) were present in pancreatic islets of healthy rats, distinguished clearly into the pancreatic α and β cells, respectively. Although streptozotocin-induced diabetic rats suffered from remarkable impairment of pancreatic islets, the localization and deposit levels of the Glycer- and MGO-AGEs were not altered in the remaining α and β cells. Remarkably, the MGO-AGEs in pancreatic β cells were localized into the insulin-secretory granules. These results suggest that the cell-specific localization of AGEs-modified proteins are presence generally in healthy peripheral tissues, involved in physiological intracellular roles, such as a post-translational modulator contributing to the secretory and/or maturational functions of insulin.

    DOI: 10.1016/j.jphs.2017.07.005

    Web of Science

    PubMed

    researchmap

  • Anti-high mobility group box-1 (HMGB1) antibody inhibits hemorrhage-induced brain injury and improved neurological deficits in rats. 国際誌

    Dengli Wang, Keyue Liu, Hidenori Wake, Kiyoshi Teshigawara, Shuji Mori, Masahiro Nishibori

    Scientific reports   7   46243 - 46243   2017年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:NATURE PUBLISHING GROUP  

    As one of the most lethal stroke subtypes, intracerebral hemorrhage (ICH) is acknowledged as a serious clinical problem lacking effective treatment. Available evidence from preclinical and clinical studies suggests that inflammatory mechanisms are involved in the progression of ICH-induced secondary brain injury. High mobility group box-1 (HMGB1) is a ubiquitous and abundant nonhistone DNA-binding protein, and is also an important proinflammatory molecule once released into the extracellular space from the nuclei. Here, we show that treatment with neutralizing anti-HMGB1 mAb (1 mg/kg, i.v. twice) remarkably ameliorated ICH-injury induced by local injection of collagenase IV in the striatum of rats. Administration of anti-HMGB1 mAb inhibited the release of HMGB1 into the extracellular space in the peri-hematomal region, reduced serum HMGB1 levels and decreased brain edema by protecting blood-brain barrier integrity, in association with decreased activated microglia and the expression of inflammation-related factors at 24 h after ICH. Consequently, anti-HMGB1 mAb reduced the oxidative stress and improved the behavioral performance of rats. These results strongly indicate that HMGB1 plays a critical role in the development of ICH-induced secondary injury through the amplification of plural inflammatory responses. Intravenous injection of neutralizing anti-HMGB1 mAb has potential as a novel therapeutic strategy for ICH.

    DOI: 10.1038/srep46243

    Web of Science

    PubMed

    researchmap

  • Anti-HMGB1 therapy for intracerebral hemorrhage

    Dengli Wang, Katsuyaku Ryu, Hidenori Wake, Kiyoshi Teshigawara, Shuji Mori, Masahiro Nishibori

    JOURNAL OF PHARMACOLOGICAL SCIENCES   133 ( 3 )   S98 - S98   2017年3月

     詳細を見る

    記述言語:英語   出版者・発行元:JAPANESE PHARMACOLOGICAL SOC  

    Web of Science

    researchmap

  • Anti-high mobility group box-1 (HMGB1) antibody attenuates delayed cerebral vasospasm and brain injury after subarachnoid hemorrhage in rats. 国際誌

    Jun Haruma, Kiyoshi Teshigawara, Tomohito Hishikawa, Dengli Wang, Keyue Liu, Hidenori Wake, Shuji Mori, Hideo Kohka Takahashi, Kenji Sugiu, Isao Date, Masahiro Nishibori

    Scientific reports   6   37755 - 37755   2016年11月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:NATURE PUBLISHING GROUP  

    Although delayed cerebral vasospasm (DCV) following subarachnoid hemorrhage (SAH) is closely related to the progression of brain damage, little is known about the molecular mechanism underlying its development. High mobility group box-1 (HMGB1) plays an important role as an initial inflammatory mediator in SAH. In this study, an SAH rat model was employed to evaluate the effects of anti-HMGB1 monoclonal antibody (mAb) on DCV after SAH. A vasoconstriction of the basilar artery (BA) associated with a reduction of nuclear HMGB1 and its translocation in vascular smooth muscle cells were observed in SAH rats, and anti-HMGB1 mAb administration significantly suppressed these effects. Up-regulations of inflammation-related molecules and vasoconstriction-mediating receptors in the BA of SAH rats were inhibited by anti-HMGB1 mAb treatment. Anti-HMGB1 mAb attenuated the enhanced vasocontractile response to thrombin of the isolated BA from SAH rats and prevented activation of cerebrocortical microglia. Moreover, locomotor activity and weight loss recovery were also enhanced by anti-HMGB1 mAb administration. The vasocontractile response of the BA under SAH may be induced by events that are downstream of responses to HMGB1-induced inflammation and inhibited by anti-HMGB1 mAb. Anti-HMGB1 mAb treatment may provide a novel therapeutic strategy for DCV and early brain injury after SAH.

    DOI: 10.1038/srep37755

    Web of Science

    PubMed

    researchmap

  • Soluble form of the receptor for advanced glycation end-products attenuates inflammatory pathogenesis in a rat model of lipopolysaccharide-induced lung injury.

    Yasuhisa Izushi, Kiyoshi Teshigawara, Keyue Liu, Dengli Wang, Hidenori Wake, Katsuyoshi Takata, Tadashi Yoshino, Hideo Kohka Takahashi, Shuji Mori, Masahiro Nishibori

    Journal of pharmacological sciences   130 ( 4 )   226 - 34   2016年4月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)   出版者・発行元:JAPANESE PHARMACOLOGICAL SOC  

    Acute respiratory distress syndrome (ARDS) is a severe respiratory failure caused by acute lung inflammation. Recently, the receptor for advanced glycation end-products (RAGE) has attracted attention in the lung inflammatory response. However, the function of soluble form of RAGE (sRAGE), which is composed of an extracellular domain of RAGE, in ARDS remains elusive. Therefore, we investigated the dynamics of pulmonary sRAGE and the effects of exogenous recombinant human sRAGE (rsRAGE) under intratracheal lipopolysaccharide (LPS)-induced lung inflammation. Our result revealed that RAGE was highly expressed on the alveolar type I epithelial cells in the healthy rat lung including sRAGE isoform sized 45 kDa. Under LPS-induced injured lung, the release of sRAGE into the alveolar space was increased, whereas the expression of RAGE was decreased with alveolar disruption. Treatment of the injured lung with rsRAGE significantly suppressed the lung edema, the neutrophils infiltration, the release of high mobility group box-1 (HMGB1), and the expressions of TNF-α, IL-1β and iNOS. These results suggest that the alveolar release of sRAGE may play a protective role against HMGB1 as well as exogenous pathogen-associated molecular patterns. Supplementary therapy with sRAGE may be an effective therapeutic strategy for ARDS.

    DOI: 10.1016/j.jphs.2016.02.005

    Web of Science

    PubMed

    researchmap

  • Anti-HMGB1 therapy for intracerebral hemorrhage

    Dengli Wang, Keyue Liu, Hidenori Wake, Kiyoshi Teshigawara, Shuji Mori, Masahiro Nishibori

    JOURNAL OF PHARMACOLOGICAL SCIENCES   130 ( 3 )   S88 - S88   2016年3月

     詳細を見る

    記述言語:英語   出版者・発行元:JAPANESE PHARMACOLOGICAL SOC  

    Web of Science

    researchmap

  • Anti-HMGB1 therapy for intracerebral hemorrhage and vasospasm after subarachnoid hemorrhage

    Dengli Wang, Katsuyaku Ryu, Masahiro Nishibori

    JOURNAL OF PHARMACOLOGICAL SCIENCES   128 ( 3 )   S39 - S39   2015年7月

     詳細を見る

    記述言語:英語   出版者・発行元:JAPANESE PHARMACOLOGICAL SOC  

    Web of Science

    researchmap

  • Contamination of Phenylobacterium in several human and murine cell cultures. 国際誌

    Tianqing Xiong, Dengli Wang, Baihong Tan, Wenhua Jiang, Gaoyue Dai, Hui Zhao, Shulei Li, Yanchao Li

    Wei sheng wu xue bao = Acta microbiologica Sinica   55 ( 2 )   176 - 86   2015年2月

     詳細を見る

    記述言語:英語   掲載種別:研究論文(学術雑誌)  

    OBJECTIVE: To identify and characterize an unknown microorganism causing contamination in several mammalian cell cultures. METHODS: This bacterium was identified by 16S rRNA sequencing and studied by DAPI and DiOC6 (3) staining, Gram staining, acid-fast staining, and electron microscopy. The isolated bacterium was also used to infect host cells to observe antibiotic effectiveness and its relationship with host cells. RESULTS: The 16S rRNA sequence analysis shows that this rod-shaped microorganism belongs to the family Caulobacteraceae, class Alphaproteobacteria, and was most closely related to Phenylobacterium zucineum HLK1T strain. The bacterium collected in the "swimming" stage was Gram staining negative, but Gram staining positive in the "sessile" stage. Under the electron microscope both flagellated and non-flagellated types were found. So far, no antibiotics were effective to inhibit this microorganism. The contamination with this bacterium frequently led to failed resuscitation of thawed cells. We found that the cells resuscitated with the used culture supernatants were increased in number by 3-4 folds as compared to those resuscitated with freshly prepared media. CONCLUSION: Phenylobacterium may have a dimorphic life cycle including a swimming stage and a sessile stalked stage.

    PubMed

    researchmap

▼全件表示

MISC

  • Butyrate誘起結腸過敏におけるマクロファージおよび腸管グリア由来HMGB1の役割

    佐々木花菜, SHIN Eunkyung, 野中結, 坪田真帆, WANG Dengli, 西堀正洋, 川畑篤史

    日本薬学会年会要旨集(Web)   142nd   2022年

     詳細を見る

  • オキサリプラチン誘発性末梢神経障害(OIPN)に対する抗血小板薬の予防効果:OPINへの血小板由来HMGB1の関与について

    川畑篤史, 岸本彩野, 堂本莉紗, 松永浩明, 松本亜紗菜, 坪田真帆, 関口富美子, WANG Dengli, 西堀正洋

    Pain Research   37 ( 4 )   2022年

     詳細を見る

  • 活性化プロテインCは神経障害性疼痛を抑制する:Proteinase-activated receptor-1(PAR1)の関与について

    圓尾賢悟, 池田裕哉, 坪田真帆, 王登莉, 西堀正洋, 南達郎, 伊藤彰敏, 川畑篤史

    生体機能と創薬シンポジウム要旨集   2022   2022年

     詳細を見る

  • Oxaliplatin誘起末梢神経障害には血小板由来HMGB1が関与する

    岸本彩野, 堂本莉紗, 松永浩明, 松本亜紗菜, 坪田真帆, 関口富美子, 王登莉, 西堀正洋, 川畑篤史

    日本薬理学雑誌   157 ( Supplement )   2022年

     詳細を見る

  • 抗がん剤vincristineおよびbortezomibにより誘起される末梢神経障害に関与するマクロファージからのHMGB1遊離メカニズムの相違

    青木葉優衣, 谷津健太, 池田裕哉, 関口富美子, 坪田真帆, WANG Dengli, 西堀正洋, 川畑篤史

    日本薬学会年会要旨集(Web)   142nd   2022年

     詳細を見る

  • TLR2はAGE2刺激による細胞増殖を抑制する

    豊村 隆男, 渡邊 政博, 和氣 秀徳, 逢坂 大樹, 王 登莉, 高橋 英夫, 西堀 正洋, 森 秀治

    日本薬学会年会要旨集   141年会   28P02 - 189   2021年3月

     詳細を見る

    記述言語:日本語   出版者・発行元:(公社)日本薬学会  

    researchmap

  • 非ヒト霊長類脳におけるヒト化a-HMGB1 mAbによる脳内出血の治療

    王登莉, 喬寒棟, 逢坂大樹, 西堀正洋

    日本薬理学雑誌   156 ( Supplement )   2021年

     詳細を見る

  • ウサギを用いた脊髄虚血再灌流障害モデルに対する抗HMGB1抗体の有用性の検討

    村岡玄哉, 劉克約, 喬寒棟, 逢坂大樹, 王登莉, 藤井泰宏, 笠原真悟, 西堀正洋

    日本胸部外科学会定期学術集会(Web)   74th   2021年

     詳細を見る

  • グリコールアルデヒド由来AGEsによる細胞増殖機構

    豊村隆男, 渡邊政博, 和氣秀徳, 西中崇, 逢坂大樹, 王登莉, 高橋英夫, 西堀正洋, 森秀治

    日本生化学会大会(Web)   94th   2021年

     詳細を見る

  • 終末糖化産物AGE2による細胞増殖効果とTLR2による制御

    豊村隆男, 渡邊政博, 和氣秀徳, 逢坂大樹, 王登莉, 高橋英夫, 西堀正洋, 森秀治

    日本生化学会大会(Web)   93rd   2020年

     詳細を見る

  • TLR2によるAGE2依存的な細胞増殖制御

    豊村隆男, 渡邊政博, 和氣秀徳, 逢坂大樹, 王登莉, 高橋英夫, 西堀正洋, 森秀治

    日本分子生物学会年会プログラム・要旨集(Web)   43rd   2020年

     詳細を見る

  • The effect of anti-HMGB1 antibody on collagenase-induced intracerebral hemorrhage in rats

    Katsuyaku Ryu, Dengli Wang, Hidenori Wake, Kiyoshi Teshigawara, Hideo Takahashi, Shuji Mori, Masahiro Nishibori

    JOURNAL OF PHARMACOLOGICAL SCIENCES   124   189P - 189P   2014年

     詳細を見る

    記述言語:英語   掲載種別:研究発表ペーパー・要旨(国際会議)   出版者・発行元:JAPANESE PHARMACOLOGICAL SOC  

    Web of Science

    researchmap

▼全件表示

講演・口頭発表等

  • 抗HMGB1抗体による脳内出血の治療:霊長類マーモセットを用いた検討

    王登莉

    第36回 創薬・薬理フォーラム岡山  2021年12月25日  広島大学薬効解析科学教室

     詳細を見る

    開催年月日: 2021年12月25日

    記述言語:英語   会議種別:口頭発表(一般)  

    国名:日本国  

  • The therapeutic effects of humanized anti-HMGB1 mAb on intracerebral hemorrhage-induced brain injury in marmoset

    王登莉

    第44回日本神経科学学会  2021年7月31日 

     詳細を見る

    開催年月日: 2021年7月28日 - 2021年7月31日

    記述言語:英語   会議種別:ポスター発表  

    researchmap

  • The therapeutic effects of anti-HMGB1 mAb on subdural hematoma-induced brain injury in rats

    王 登莉

    第43回日本神経科学学会  2020年7月29日 

     詳細を見る

    開催年月日: 2020年7月28日 - 2020年8月1日

    記述言語:英語   会議種別:口頭発表(一般)  

    researchmap

  • Effects of anti-HMGB1 mAb in various models of brain injury 招待

    王登莉

    Soochow University Forum for Overseas High-level Talents  2019年12月27日 

     詳細を見る

    開催年月日: 2019年12月26日 - 2019年12月28日

    記述言語:英語   会議種別:口頭発表(招待・特別)  

    researchmap

  • Anti-HMGB1 therapy for intracerebral and subdural hemorrhage-induced brain injury in rats

    王登莉

    9th International DAMPs and Alarmins Symposium  2019年11月7日 

     詳細を見る

    開催年月日: 2019年11月6日 - 2019年11月8日

    記述言語:英語   会議種別:ポスター発表  

    researchmap

  • Anti-HMGB1 mAb ameliorates intracerebral hemorrhage-induced brain injury in rats

    王登莉

    第41回日本神経科学大会  2018年7月27日 

     詳細を見る

    開催年月日: 2018年7月26日 - 2018年7月29日

    記述言語:英語   会議種別:口頭発表(一般)  

    researchmap

  • Anti-HMGB1 mAb therapy for intracerebral hemorrhage-induced brain injury in rats

    王登莉

    第18回国際薬理学・臨床薬理学会議  2018年7月3日 

     詳細を見る

    開催年月日: 2018年7月1日 - 2018年7月6日

    記述言語:英語   会議種別:口頭発表(一般)  

    researchmap

  • Treatment of intracerebral hemorrhage with humanized a-HMGB1 mAb in non-human primate brain

    王登莉

    第139回日本薬理学会近畿部会  2021年6月26日 

     詳細を見る

    記述言語:英語   会議種別:口頭発表(一般)  

    researchmap

  • HMGB1 Translocation in Neurons after Ischemic Insult: Subcellular Localization in Mitochondria and Peroxisomes

    王登莉

    第137回日本薬理学会近畿部会  2020年6月20日 

     詳細を見る

    記述言語:英語   会議種別:ポスター発表  

    researchmap

  • Anti-HMGB1 mAb therapy for intracerebral and subdural hemorrhage in rats

    王登莉

    第93回日本薬理学会年会  2020年3月 

     詳細を見る

    記述言語:英語   会議種別:口頭発表(一般)  

    researchmap

  • Anti-HMGB1 therapy for intracerebral and subdural hemorrhage-induced brain injury in rats

    王登莉

    第31回創薬・薬理学フォーラム  2019年7月 

     詳細を見る

    記述言語:英語   会議種別:口頭発表(一般)  

    researchmap

  • Anti-HMGB1 mAb ameliorates intracerebral hemorrhage-induced brain injury in rats

    王登莉

    第29回創薬・薬理学フォーラム  2018年7月 

     詳細を見る

    記述言語:英語   会議種別:口頭発表(一般)  

    researchmap

▼全件表示

受賞

  • 優秀演題賞

    2016年6月   第37回日本炎症・再生医学会  

     詳細を見る

  • 国際交流奨励賞

    2015年3月   公益信託岡山ロータリークラブ国際交流振興基金  

    王登莉

     詳細を見る

共同研究・競争的資金等の研究

  • がん増悪因子としての溶血とヘモペキシンの新規活性に着目した治療戦略検証と創薬

    研究課題/領域番号:22K07253  2022年04月 - 2025年03月

    日本学術振興会  科学研究費助成事業  基盤研究(C)

    逢坂 大樹, 王 登莉, 細野 祥之

      詳細を見る

    配分額:4160000円 ( 直接経費:3200000円 、 間接経費:960000円 )

    researchmap

  • HMGB1-HRGバランスに着目した川崎病新規治療薬の開発

    研究課題/領域番号:21K07822  2021年04月 - 2024年03月

    日本学術振興会  科学研究費助成事業  基盤研究(C)

    馬場 健児, 平井 健太, 西堀 正洋, 逢坂 大樹, 王 登莉

      詳細を見る

    配分額:4160000円 ( 直接経費:3200000円 、 間接経費:960000円 )

    2021年度は、川崎病急性期患者における血中HMGB1、HRG濃度の測定において大きな成果が得られた。予定していた人数で川崎病急性期患者における治療前と初回治療後の血漿検体採取が終了し、コントロールとして健常者の検体と血中HMGB1、HRG濃度の比較を行ったところ、川崎病患者では健常者に比べて血中HMGB1が有意に高値、血中HRGが有意に低値であった。さらに川崎病患者を初回治療反応群、不応群に分け、治療前後の血中HMGB1、HRG濃度の変化についても詳細な解析を行い、論文投稿の準備を進めている。
    また川崎病モデルマウスの実験に関しては、Lactobacillus cell wall extract(LCWE)を作成し、マウスに腹腔内投与することで、川崎病様の冠動脈炎が惹起されることを確認した。しかし個体によって冠動脈炎の程度にばらつきが大きかったため、同様に川崎病冠動脈炎を惹起するCandida albicans water soluble fraction(CAWS)を用いるモデルについても検討を行った。CAWSを作成してマウスに腹腔内投与したところ、LCWEよりも安定してモデル作成ができることが明らかとなったため、以後の検討ではCAWSを腹腔内投与するモデルを使用することとした。

    researchmap

  • 抗HMGB1モノクローナル抗体による大動脈手術後対麻痺予防効果の検討

    研究課題/領域番号:21K08823  2021年04月 - 2024年03月

    日本学術振興会  科学研究費助成事業  基盤研究(C)

    村岡 玄哉, 大澤 晋, 藤井 泰宏, 王 登莉, 西堀 正洋, 逢坂 大樹, 笠原 真悟

      詳細を見る

    配分額:4030000円 ( 直接経費:3100000円 、 間接経費:930000円 )

    胸部下行・胸腹部の大動脈瘤手術は大動脈ステントグラフトの発達に伴いハイリスク患者
    でも積極治療されるようになり、急激に患者数が増加している。本邦での最新の胸部・胸腹部大動脈手術数は35,427例(2015年1月~2016年2月)である。一方、これらの疾患群の死亡率は平均7.3%と高く、生命予後に大きく関わる非常に重大な合併症として、脊髄虚血による対麻痺が挙げられる。対麻痺は術式にもよるが、3.4~10.4% の確率で発生しており、かなりの数の患者が対麻痺を発症している。(日心外科誌48巻1号: 18-24, 2019)。この発生率は、既存の脊髄保護法(Naroxone投与、冷却、脊髄ドレナージ、Adamkiewics動脈再建、ステロイド等)を適宜行った上での数値であり、新たな対策・治療法開発は喫緊の課題である。我々はこれまで、岡山大学薬理学の西堀らが独自に開発した抗HMGB1(high mobility group box 1)抗体を用い、脳虚血、くも膜下出血、硬膜下出血、脳外傷といった中枢神経疾患動物モデルでその脳障害予防効果を証明してきた。そこで、心臓血管外科手術における脊髄保護にも有用ではないかと考え、ウサギの脊髄虚血モデル作成のPreliminary Study を施行後、本研究を計画した。本研究の目的は、NSC 34 Cell を分化させた、Motor Neuron Like Cellとうさぎ脊髄虚血モデルを用いて、HMGB1抗体のMotor Neuron に対する細胞死抑制効果、酸化ストレス抑制効果を実証し、臨床応用へ繋げる事である。本年度は、計画している実験のうち、ウサギを用いた実験を行い、細胞を使用する実験は来年以降に行うこととした。ウサギの脊髄虚血モデルを用いた抗HMGB-1抗体の脊髄麻痺予防効果実証実験R3年度予定分を施行した。

    researchmap

  • 霊長類マーモセットを用いた脳内出血治療法の開発と基盤となるHMGB1動態の解析

    研究課題/領域番号:20K17930  2020年04月 - 2024年03月

    日本学術振興会  科学研究費助成事業  若手研究

    王 登莉

      詳細を見る

    配分額:4160000円 ( 直接経費:3200000円 、 間接経費:960000円 )

    令和2年度の研究計画では霊長類マーモセット脳出血モテルにおけるヒト化抗 HMGB1 抗体の効果の確認を目的とした。 マーモセットの右基底核にコラケナーゼIVを定位的に注入することで脳出血モデルを作成した。脳出血モデル作成後10分間にヒト化抗 HMGB1抗体を尾静脈から投与した。対照動物には、ヒトIgGを投与した。2時間、6時間、1日、2日、3日、6日、12日後に採血し、体重を測定した。神経学的評価は、経時的にgrip strengthテストを実施した。脳損傷に対する抗HMGB1 の効果を測定するために、脳出血後6、12 日後の CT 像を撮像し脳組織損傷範囲を評価した。12日後に脳を取り出しHE染色や免疫染色をした。その結果、抗HMGB1抗体の投与によって、血漿中HMGB1と酸化ストレスマーカーである4-HNEの有意に抑制を示した。出血後から12日間、対照動物の体重がずっと減少することが明瞭であったが、抗HMGB1抗体の投与によって3日間から回復し6日間で出血前の体重に戻った。grip strengthテストによる評価で、運動麻痺神経症状も、抗HMGB1抗体で高い改善効果を示した。また、脳出血12日C T像から見ると、抗HMGB1抗体の投与によって脳組織損傷範囲は減少した。

    researchmap

  • 敗血症関連DAMPs拮抗因子HRGの血管保護作用機序の解明

    研究課題/領域番号:20K07290  2020年04月 - 2023年03月

    日本学術振興会  科学研究費助成事業  基盤研究(C)

    和氣 秀徳, 勅使川原 匡, 王 登莉

      詳細を見る

    配分額:4420000円 ( 直接経費:3400000円 、 間接経費:1020000円 )

    HRGのフェロトーシス細胞死への関与を明らかにするために、血管内皮細胞株EA.hy926細胞へ2価鉄もしくは3価鉄で刺激を行い、鉄による細胞障害性をMTSアッセイを用いて評価した。2価の鉄は鉄を添加していない群や3価の鉄を添加した群と比較して、著しい細胞活動の低下を示し、HRGはこの低下をほぼ完全に抑制した。また細胞非透過性の核染色剤SytoxOrangeを用いて細胞膜完全性の評価を行ったところ、2価の鉄は鉄を添加していない群や3価の鉄を添加した群と比較して、細胞膜が障害されており、HRGはこの細胞膜障害を抑制した。さらに、細胞膜脂質過酸化状態を過酸化脂質蛍光染色剤で評価したところ、2価鉄刺激により細胞膜の酸化が引き起こされ、HRGはこれを抑制することを明らかにした。つまり、HRGは2価鉄による細胞膜酸化障害を抑制することで、細胞を保護する活性を有することが明らかとなった。
    HRGのパイロトーシス経路への関与に関しては、以前からパイロトーシスを誘導することで知られているLPSにより、パイロトーシスへの関与が疑われるHMGB1の血管内皮細胞核内から細胞外へのトランスロケーションを誘導することを明らかにしていたが、HRGはCLEC1Aを介して、このHMGB1トランスロケーションを阻害することを新たに示した。
    HRGの血管内皮細胞表面上のグリコカリックスの状態に対する影響に関しては、マウスの血管内皮細胞表面グリコカリックスをを蛍光標識レクチンで染色し、インビボイメージングにより、その状態を評価した。LPSのi.v.投与により、蛍光標識レクチンで染色されない血管が増加し、HRG KOマウスではその割合がさらに増加することから、血中のHRGはLPSによるグリコカリックスの剥離を抑制する効果を有することが示唆された。

    researchmap

  • 組織障害性HMGB1に着眼した肺虚血再灌流障害に対する新規戦略の確立

    研究課題/領域番号:20K09176  2020年04月 - 2023年03月

    日本学術振興会  科学研究費助成事業  基盤研究(C)

    大谷 真二, 山根 正修, 豊岡 伸一, 杉本 誠一郎, 王 登莉, 西堀 正洋, 岡崎 幹生, 三好 健太郎, 阪口 政清

      詳細を見る

    配分額:4290000円 ( 直接経費:3300000円 、 間接経費:990000円 )

    ①マウス肺門クランプモデルにおけるHMGB1の動態:HMGB1は通常核内に限局しているタンパクである。マウス左肺門クランプモデルにおいて、再還流後2時間までのHMGB1の血中濃度を測定したところ、HMGB1が時系列に沿って上昇することが確認された。また組織の免疫染色において、虚血再還流障害を加えた群では、HMGB1が核内よりむしろ細胞質で強く染色され、細胞障害性の刺激により核内から細胞質へ移動している様子が確認された。この現象はHMGB1抗体を投与することで抑制されたことから細胞障害によりHMGB1には自己分泌経路が出現し抗体投与によってその経路を停止させることができるのではないか、と考えられた。
    ②抗体投与による虚血再還流障害の抑制:HMGB1抗体を投与する事による虚血再還流障害の抑制効果を調べた。肺機能、組織障害の改善を生理学的、組織学的に確認することができた。
    ③抗体投与による抗炎症効果:HMGB1抗体を投与するとサイトカインの産生が低下することが確認された。HMGB1はRAGEやTLRといったレセプターのリガンドであり、MAPKの経路を介しサイトカイン産生を行っているが、HMGB1抗体によりMAPKの経路が抑制されることが示された。
    ④抗体投与によるアポトーシスの抑制:肺組織の細胞死を定量するため組織でのアポトーシスを検索した。虚血性再還流障害によるアポトーシスが抗体投与により抑制されていることが確認された。

    researchmap

  • バイオセンサー分子としてのHMGB1の動態と多機能性解析

    研究課題/領域番号:19H03408  2019年04月 - 2022年03月

    日本学術振興会  科学研究費助成事業  基盤研究(B)

    西堀 正洋, 阪口 政清, 逢坂 大樹, 王 登莉, 和氣 秀徳, 勅使川原 匡

      詳細を見る

    配分額:17550000円 ( 直接経費:13500000円 、 間接経費:4050000円 )

    組織損傷関連分子群(Damage-associated molecular patterns; DAMPs)の代表である核内因子High mobility group box-1 (HMGB1) は、起炎性刺激によって細胞外へ放出される。一方、申請者はHMGB1が生理的活性物質によっても、血管内皮細胞から分泌されることを明らかにし、生理機能調節に関与するバイオセンサーとしての機能に注目するに至った。すでに、HMGB1の放出反応は抗HMGB1抗体の添加で殆ど完全に抑制されることを観察し、ポジティブフィードバックの存在を推定している。本研究では、血管内皮細胞をモデル細胞として用いることにより、HMGB1の分泌過程を解明し、一連のサイトカイン産生における増幅機能、内皮細胞ホメオステーシスにおける意義、細胞質内における新規機能の全体像を明らかにする。
    平成31(令和元)年度は、トロンビン刺激あるいはヒスタミン刺激によって、培養ヒト血管内皮細胞(EA.hy926)の細胞核から細胞質を経由して細胞外へHMGB1が分泌されることを明らかにした。ヒスタミン刺激によるHMGB1の分泌は、ヒスタミンの濃度依存的に生じ、ヒスタミン添加後時間依存的に進行した。4種類のヒスタミン受容体アンタゴニストの内、H1-選択的なアンタゴニストであるd-クロルフェニラミンのみがヒスタミン刺激によるHMGB1分泌を抑制し、またH1-受容体アゴニストの2-ピリジルエチルアミンがヒスタミンの作用を模倣したことから、HMGB1トランスロケーションに関与する受容体はH1-受容体であると結論された。

    researchmap

  • HRG遺伝子欠損マウスの妊娠期表現型に基づく妊娠高血圧症候群の病態解析

    研究課題/領域番号:19K07401  2019年04月 - 2022年03月

    日本学術振興会  科学研究費助成事業  基盤研究(C)

    勅使川原 匡, 西堀 正洋, 和氣 秀徳, 王 登莉, 劉 克約

      詳細を見る

    配分額:4290000円 ( 直接経費:3300000円 、 間接経費:990000円 )

    高ヒスチジン糖タンパク(histidine-rich glycoprotein, HRG)は、分子量75kDaの血漿糖タンパクである。微小血栓形成や血管内皮細胞障害などの炎症病態に対して、HRGは、炎症惹起因子であるHMGB1の血管新生作用や好中球の過剰活性を抑制するなどの抗炎症的役割を果たす。近年、ヒト妊婦において血漿HRG量の低下が妊娠高血圧腎症の進展と相関することが報告されている。自然免疫系応答にみられる炎症性の血液・血管病態イベントは、周産期の胎盤形成・妊娠の維持においても重要な生理的現象であり、HRGの周産期生理における関与が示唆される。本研究では、妊娠高血圧症候群(HDP)の病態や周産期生理におけるHRGの役割を明らかとするために、HRG遺伝子欠損(HRG KO)マウスの妊娠期表現型や妊娠野生型C57BL/6マウスの血漿HRGの動態を解析した。
    HRG KOマウスは、野生型マウスと比べて、血中HMGB1量が恒常的に高値であった。妊娠18日目の胎盤組織は、炎症関連因子(IL-1 beta, TNF-alpha, iNOS)の発現量に変化がなかったが、血管新生因子(VEGF, PlGF)の発現量に増加がみられた。また、胎盤過形成、及び、高血圧がみとめられた。
    ヒト妊婦と同様に、妊娠野生型C57BL/6マウスにおいても血漿HRGの減少が認められた。この血漿HRGの減少は、主要賛成臓器である肝でのHRG遺伝子発現に依存しておらず、タンパクレベルでのクリアランス亢進が原因と考えられた。
    これらの結果から、抗炎症因子であるHRGが、妊娠期の胎盤形成・血圧調節に何らかの生理的役割をもつ可能性が示唆された。妊娠HRG KOマウスは、HRGによって制御される胎盤形成機序に異常を生じるHDP様病態モデルとなる可能性がある。

    researchmap

▼全件表示

 

担当授業科目

  • 先端創薬研究序論 (2023年度) 特別

  • 医学セミナー (2023年度) 第1学期

  • 医学研究インターンシップ (2023年度) 特別

  • 基礎病態演習 (2023年度) 特別

  • 総合薬理学 (2023年度) 集中

  • 薬理学 (2023年度) 特別  - その他

  • 薬理学実習 (2023年度) 特別  - その他

  • 医学セミナー (2022年度) 第1学期

  • 医学研究インターンシップ (2022年度) 特別

  • 基礎病態演習 (2022年度) 特別

  • 薬理学 (2022年度) 特別  - その他

  • 薬理学実習 (2022年度) 特別  - その他

  • 医学研究インターンシップ (2021年度) 特別

  • 薬理学 (2021年度) 特別  - その他

  • 薬理学実習 (2021年度) 特別  - その他

▼全件表示

 

社会貢献活動

  • 2023年度O-NECUSプログラムオータムセミナー

    役割:司会

    2022年11月9日

  • 中国・浙江大学医学部生 岡山大学夏期フロクラム

    役割:講師

    2018年7月 - 2019年7月

     詳細を見る

    種別:その他

    researchmap