研究者業績

小野 秀樹

オノ ヒデキ  (ONO HIDEKI)

基本情報

所属
武蔵野大学 薬学部 薬学科 教授
学位
薬学博士(東京大学)

J-GLOBAL ID
200901084214854756
researchmap会員ID
1000212505

学歴

 2

論文

 167
  • Akihiro Fukushima, Arisa Fukui, Yuki Takemura, Yasuhiro Maeda, Hideki Ono
    Journal of pharmacological sciences 136(1) 39-41 2018年1月  
    Oseltamivir has a hypothermic effect in mice when injected intraperitoneally (i.p.) and intracerebroventricularly (i.c.v.). Here we show that the hypothermia evoked by i.c.v.-oseltamivir is inhibited by non-selective dopamine receptor antagonists (sulpiride and haloperidol) and the D2-selective antagonist L-741,626, but not by D1/D5-selective and D3-selective antagonists (SCH-23390 and SB-277011-A, respectively). The hypothermic effect of i.p.-administered oseltamivir was not inhibited by sulpiride, haloperidol, L-741,626 and SCH-23390. In addition, neither sulpiride, haloperidol nor SCH-23390 blocked hypothermia evoked by i.c.v.-administered oseltamivir carboxylate (a hydrolyzed metabolite of oseltamivir). These results suggest that oseltamivir in the brain induces hypothermia through activation of dopamine D2 receptors.
  • Akihiro Fukushima, Wakana Sekiguchi, Kizuku Mamada, Yumi Tohma, Hideki Ono
    BIOLOGICAL & PHARMACEUTICAL BULLETIN 40(2) 227-233 2017年2月  査読有り
    Acetaminophen (AcAP), a widely-used antipyretic and analgesic drug, has been considered to exert its effects via central mechanisms, and many studies have demonstrated that the analgesic action of AcAP involves activation of the serotonergic system. Although the serotonergic system also plays an important role in thermoregulation, the contribution of serotonergic activity to the hypothermic effect of AcAP has remained unclear. In the present study, we examined whether the serotonergic system is involved in AcAP-induced hypothermia. In normal mice, AcAP (300 mg/kg, intraperitoneally (i.p.)) induced marked hypothermia (ca. -4 degrees C). The same dose of AcAP reduced pain response behavior in the formalin test. Pretreatment with the serotonin synthesis inhibitor DL-p-chlorophenylalanine (PCPA, 300 mg/kg/d, i.p., 5 consecutive days) substantially decreased serotonin in the brain by 70% and significantly inhibited the analgesic, but not the hypothermic action of AcAP. The same PCPA treatment significantly inhibited the hypothermia induced by the selective serotonin reuptake inhibitor fluoxetine hydrochloride (20mg/kg, i.p.) and the serotonin 5-HT2 receptor antagonist cyproheptadine hydrochloride (3 mg/kg, i.p.). The lower doses of fluoxetine hydrochloride (3 mg/kg, i.p.) and cyproheptadine hydrochloride (0.3 mg/kg, i.p.) did not affect the AcAP-induced hypothermia. These results suggest that, in comparison with its analgesic effect, the hypothermic effect of AcAP is not mediated by the serotonergic system.
  • Shohei Yamamoto, Yuma Suzuki, Hideki Ono, Kazuhiko Kume, Masahiro Ohsawa
    EUROPEAN JOURNAL OF PHARMACOLOGY 793 66-75 2016年12月  査読有り
    Cilnidipine is a dihydropyridine derivative that inhibits N-type and L-type voltage-gated Ca2+ channels (VDCCs). We recently reported that a selective N-type VDCC blocker attenuated the spinal long-term potentiation (LTP) of C-fiber-evoked field potentials recorded in the spinal dorsal horn of rats, which served as a model for examining synaptic function during central pain sensitization. In this study, we investigated the effects of cilnidipine on the changes related to neuropathic pain induced by nerve injury. Mechanical allodynia and hyperalgesia were evaluated by von Frey test and pin prick test, respectively. Spinal LTP of C-fiber-evoked field potentials were evaluated by in vivo electrophysiology. Intrathecally administrated cilnidipine attenuated mechanical allodynia and hyperalgesia in the spared nerve injury mouse model. Using in vivo electrophysiology in rats, cilnidipine (10 mu m) administered spinally inhibited the induction and maintenance of high-frequency stimulation-induced LTP of C-fiber-evoked field potentials, while basal C-fiber-evoked field potentials in naive rats were unaffected. The basal C-fiber-evoked field potentials in nerve-injured rats were strongly inhibited by cilnidipine. Treatment with a specific N-type VDCC blocker, omega o-conotoxin GVIA, which reportedly attenuates C-fiber-evoked field potentials both before and after the induction of LTP, attenuated mechanical allodynia and hyperalgesia in nerve-injured mice. By contrast, an L-type VDCC blocker, nicardipine attenuated only mechanical hyperalgesia, but not mechanical allodynia in nerve-injured mice, and also attenuated the established LTP of C-fiber-evoked field potentials in rats. These results suggested that N-type and L-type VDCC blockers may effectively alleviate the hyperalgesia and allodynia associated with neuropathic pain without affecting normal pain perception.
  • Shohei Yamamoto, Hideki Ono, Kazuhiko Kume, Masahiro Ohsawa
    JOURNAL OF PHARMACOLOGICAL SCIENCES 130(4) 189-193 2016年4月  査読有り
    Oxaliplatin (L-OHP) is a platinum-based chemotherapy drug, used in standard treatment of colorectal cancer. L-OHP frequently causes acute peripheral neuropathies. These adverse effects limit cancer therapy with L-OHP. The present study was designed to reveal the changes in sensory nerve function in L-OHP-injected rats. Mechanical static allodynia, dynamic allodynia, and cold allodynia were evaluated using the von Frey test, brush test, and acetone test, respectively. Sensory nerve fiber responsiveness was measured using a Neurometer. The fifth lumbar ventral root was sectioned to record multi-unit efferent discharges. Single intraperitoneal administration of L-OHP induced mechanical static allodynia, dynamic allodynia, and cold allodynia in Wistar/ST rats. The thresholds for paw withdrawal induced by 2000 Hz (A beta-fiber) and 5 Hz (C-fiber), but not 250 Hz (A delta-fiber) sine-wave electrical stimulation were reduced in L-OHP-treated rats. Multi-unit efferent discharges were increased by mechanical stimulation using a von Frey filament applied to the plantar surface of the hindpaw. The discharges during and after stimulation were increased in the L-OHP-treated rats. Cold stimulation, but not brush stimulation, increased the discharges in L-OHP-treated rats. These results suggest that sensitization of A beta- and C-fibers, but not A delta-fibers, contributes to the development of L-OHP-induced mechanical and cold allodynia. (C) 2015 The Authors. Production and hosting by Elsevier B.V. on behalf of Japanese Pharmacological Society. This is an open access article under the CC BY-NC-ND license.
  • Akihiro Fukushima, Kaori Chazono, Yuichi Hashimoto, Yui Iwajima, Shohei Yamamoto, Yasuhiro Maeda, Masahiro Ohsawa, Hideki Ono
    EUROPEAN JOURNAL OF PHARMACOLOGY 762(5) 275-282 2015年9月  査読有り
    Oseltamivir, an anti-influenza virus drug, induces marked hypothermia in normal mice. We have proposed that the hypothermic effect arises from inhibition of the nicotinic acetylcholine receptor function of sympathetic ganglion neurons which innervate the brown adipose tissue (a heat generator). It has been reported that local anesthetics inhibit nicotinic acetylcholine receptor function by acting on its ionic channels, and that bupropion, a nicotinic antagonist, induces hypothermia. In this study, we compared the effects of oseltamivir, procaine and bupropion on body temperature, cardiovascular function and neuromuscular transmission Intraperitoneal administration of oseltamivir (100 mg/kg), procaine (86.6 mg/kg) and bupropion (86.7 mg/kg) lowered the core body temperature of normal mice. At lower doses (10-30 mg/kg oseltamivir, 8.7-26 mg/kg procaine and bupropion), when administered subcutaneously, the three drugs antagonized the hypothermia induced by intraperitoneal injection of nicotine (1 mg/kg). In anesthetized rats, intravenous oseltamivir (30-100 mg/kg), procaine (10 mg/kg) and bupropion (10 mg/kg) induced hypotension and bradycardia. Oseltamivir alone (100 mg/kg) did not inhibit neuromuscular twitch contraction of rats, but at 3-30 mg/kg it augmented the muscle-relaxing effect of d-tubocurarine. Similar effects were observed when lower doses of procaine (10-30 mg/kg) and bupropion (3-10 mg/kg) were administered, suggesting that systemic administration of oseltamivir inhibits muscular nicotinic acetylcholine receptors. These results support the idea that the hypothermic effect of oseltamivir is due to its effects on sympathetic ganglia which innervate the brown adipose tissue, and suggest that oseltamivir may exert non-selective ion channel blocking effects like those of ester-type local anesthetics. (C) 2015 Elsevier B.V. All rights reserved.
  • Katsuhiko Muraki, Noriyuki Hatano, Hiroka Suzuki, Yukiko Muraki, Yui Iwajima, Yasuhiro Maeda, Hideki Ono
    BASIC & CLINICAL PHARMACOLOGY & TOXICOLOGY 116(2) 87-95 2015年2月  査読有り
    The effects of oseltamivir, a neuraminidase inhibitor, were tested on the function of neuronal nicotinic acetylcholine receptors (nAChRs) in a neuroblastoma cell line IMR32 derived from human peripheral neurons and on recombinant human 34 nAChRs expressed in HEK cells. IMR32 cells predominately express 34 nAChRs. Nicotine (nic, 30m)-evoked currents recorded at -90mV in IMR32 cells using the whole-cell patch clamp technique were reversibly blocked by oseltamivir in a concentration-dependent manner. In contrast, an active metabolite of oseltamivir, oseltamivir carboxylate (OC) at 30m had little effect on the nic-evoked currents. Oseltamivir also blocked nic-evoked currents derived from HEK cells with recombinant 34 nAChRs. This blockade was voltage-dependent with 10, 30 and 100 m oseltamivir inhibiting similar to 50% at -100, -60 and -40mV, respectively. Non-inactivating currents in IMR32 cells and in HEK cells with 34 nAChRs, which were evoked by an endogenous nicotinic agonist, ACh (5m), were reversibly blocked by oseltamivir. These data demonstrate that oseltamivir blocks nAChRs, presumably via binding to a site in the channel pore.
  • 渡邊美智留, 三田恭平, 中村春世, 田中恒明, 三原潔, 小野秀樹
    医療薬学 40(12) 726-733 2014年12月  査読有り
    Pregabalin, a useful drug for neuropathic pain, has a high incidence of dizziness and somnolence as side effects. In the present study, the incidence of both side effects and the risk factors were retrospectively investigated in hospitalized patients who were administered pregabalin after their admission. In 65 patients (median 68 years old), 34 cancer patients and 18 opioid users were included. Items studied were cancer/non-cancer, opioid user/non-user, fall/nonfall, age, sex, weight, daily dose of opioids, the number of the drugs that may cause dizziness and somnolence, daily dose of pregabalin, and the ratio of creatinine clearance to daily dose of pregabalin. Fourteen (21.5%) and 21 (23.3%) patients developed dizziness and somnolence, respectively, and 4 (6.1%) patients developed fall. In the case of opioid combination, 7 (38.9%) and 10 (55.6%) patients developed dizziness and somnolence, respectively. A logistic-regression analysis showed that opioid use is a significant augmenting risk factor for dizziness (P = 0.026) and somnolence (P = 0.016) of pregabalin. The ratios of daily dose of pregabalin to creatinine clearance did not show any relation to the incidence of dizziness and somnolence; both side effects were observed in some patients whose renal functions were normal. It is suggested that attention is necessary to the incidence of dizziness and somnolence regardless of renal function, and that particular attention is required when opioids, which have similar side effects, are combined with pregabalin.
  • Muraki K, Ono H
    Receptors & Clinical Investigation 1(6) e360 2014年12月  査読有り招待有り
  • Orie Tomoyose, Daisuke Kodama, Hideki Ono, Mitsuo Tanabe
    JOURNAL OF PHARMACOLOGICAL SCIENCES 126(2) 136-145 2014年10月  査読有り
    Fluvoxamine, a selective serotonin (5-HT) reuptake inhibitor, has been shown to exert analgesic effects in humans and laboratory animals. However, its effects on spinal nociceptive synaptic transmission have not been fully characterized. Here, whole-cell recordings were made from dorsal horn neurons in spinal slices with attached dorsal roots from adult mice, and the effects of fluvoxamine on monosynaptic A-fiber- and C-fiber-mediated excitatory postsynaptic currents (EPSCs) evoked in response to electrical stimulation of a dorsal root were studied. Fluvoxamine (10 - 100 mu M) concentration-dependently suppressed both monosynaptic A-fiber- and C-fiber-mediated EPSCs, which were attenuated by the selective 5-HT1A receptor antagonist WAY100635. In the presence of the selective 5-HT3 receptor antagonist tropisetron, fluvoxamine hardly suppressed A-fiber-mediated EPSCs, whereas its inhibitory effect on C-fiber-mediated EPSCs was not affected. Although fluvoxamine increased the paired-pulse ratio of A-fiber-mediated EPSCs, it increased the frequency of spontaneous and miniature EPSCs (sEPSCs and mEPSCs). Since sEPSCs and mEPSCs appeared to arise largely from spinal interneurons, we then recorded strontium-evoked asynchronous events occurring after A-fiber stimulation, whose frequency was reduced by fluvoxamine. These results suggest that fluvoxamine reduces excitatory synaptic transmission from primary afferent fibers via presynaptic mechanisms involving 5-HT1A and/or 5-HT3 receptors, which may contribute to its analgesic effects.
  • Masahiro Ohsawa, Saki Otake, Tomoyasu Murakami, Shohei Yamamoto, Toshiaki Makino, Hideki Ono
    JOURNAL OF PHARMACOLOGICAL SCIENCES 125(3) 292-299 2014年7月  査読有り
    Oxaliplatin, a platinum-based chemotherapy drug, frequently causes acute and chronic peripheral neuropathies including mechanical hyperalgesia. These adverse effects hinder anticancer therapy with the drug. In this study, we examined several drugs that might prevent oxaliplatin-induced peripheral neuropathy. Single intraperitoneal (i.p.) injection of oxaliplatin (10 mg/kg) induced cold allodynia (acetone test) and mechanical hyperalgesia (von Frey test). Gabapentin, but not simvastatin and atorvastatin, prevented oxaliplatin-induced mechanical hyperalgesia without affecting cold allodynia. Moreover, oxaliplatin caused phosphorylation of cofilin protein in the spinal cord, which has been shown to be involved in the neuropathic hyperalgesia. This increased phosphorylation of cofilin was also attenuated by gabapentin treatment. These results suggest that gabapentin is useful for relieving oxaliplatin-induced mechanical hyperalgesia and that the pathogenic mechanisms of cold allodynia and mechanical hyperalgesia differ.
  • 大澤 匡弘, 山本 昇平, 小野 秀樹
    YAKUGAKU ZASSHI 134(3) 387-395 2014年  査読有り
  • M. Ohsawa, Y. Miyabe, H. Katsu, S. Yamamoto, H. Ono
    NEUROSCIENCE 247 65-74 2013年9月  査読有り
    Lysophosphatidic acid (LPA) has been considered one of the molecular culprits for neuropathic pain. Understanding how LPA changes the function of primary afferent fibers might be an essential step for clarifying the pathogenesis of neuropathic pain. The present study was designed to identify the primary afferent fibers (A beta, A delta, or C) participating in LPA-induced allodynia in ddY mice. Mechanical allodynia and thermal hyperalgesia were evaluated by the von Frey filament test and thermal paw withdrawal test, respectively. Sensory nerve fiber responsiveness was measured using a Neurometer. Daily repeated intrathecal treatment with LPA led to a decrease in the mechanical, but not thermal nociceptive threshold, and a reduction in the threshold for paw withdrawal induced by 2000-Hz (A beta fiber) and 250-Hz (A delta fiber), but not 5-Hz (C fiber) sine-wave electrical stimulation. When the transient receptor potential cation channel subfamily V member 1 (TRPV1) receptor agonist resiniferatoxin (RTX) was administered subcutaneously before the start of LPA treatment, LPA-induced mechanical allodynia and A beta and A delta fiber hypersensitivity demonstrated by neurometry were not affected, indicating that TRPV1-expressing nerve fibers (possibly C fibers) might not be essential for LPA-induced allodynia. LPA-induced allodynia was reversed by treatment with RTX at 7 days after the start of LPA treatment. Expression of TRPV1 on myelinated nerve fibers after repeated intrathecal LPA treatment was observed in the dorsal root ganglion. These results suggest that sensitization of A beta and A delta fibers, but not C fibers, contributes to the development of intrathecally administered LPA-induced mechanical allodynia. Moreover, increased or newly expressed TRPV1 receptors in A beta and A delta fibers are considered to be involved in the maintenance of LPA-induced allodynia. (C) 2013 IBRO. Published by Elsevier Ltd. All rights reserved.
  • Hideki Ono, Yui Iwajima, Yuko Nagano, Kaori Chazono, Yasuhiro Maeda, Masahiro Ohsawa, Shohei Yamamoto
    Basic and Clinical Pharmacology and Toxicology 113(1) 25-30 2013年7月  査読有り
    Oseltamivir, an anti-influenza virus drug, has strong antipyretic effects in mice (Biological and Pharmaceutical Bulletin, 31, 2008, 638) and patients with influenza. In addition, hypothermia has been reported as an adverse event. The prodrug oseltamivir is converted to oseltamivir carboxylate (OC), an active metabolite of influenza virus neuraminidase. In this study, core body temperature was measured in mice, and oseltamivir and OC were administered intracerebroventricularly (i.c.v.) or intraperitoneally (i.p). Low i.c.v. doses of oseltamivir and OC dose-dependently produced hypothermia. Zanamivir (i.c.v.), another neuraminidase inhibitor, did not produce hypothermia. These results suggested that the hypothermic effects of oseltamivir (i.p. and i.c.v.) and OC (i.c.v.) are not due to neuraminidase inhibition. OC (i.p.) did not lower body temperature. Although mecamylamine (i.c.v.) blocked the hypothermic effect of nicotine-administered i.c.v., the hypothermic effects of oseltamivir and OC (i.c.v.) were not blocked by mecamylamine (i.c.v.). The effect of oseltamivir (i.p.) was markedly increased by s.c.-pre-administered mecamylamine and also hexamethonium, a peripherally acting ganglionic blocker, suggesting their potentiating interaction at peripheral sites. The hypothermic effect of nicotine (i.c.v.) was decreased by lower doses of oseltamivir (i.c.v.), suggesting the anti-nicotinic action of oseltamivir. These results suggest that oseltamivir (i.p.) causes hypothermia through depression of sympathetic temperature regulatory mechanisms via inhibition of nicotinic receptor function and through unknown central mechanisms. © 2013 Nordic Pharmacological Society. Published by John Wiley &amp Sons Ltd.
  • 大澤匡弘, 藤沢佳史, 勝弘毅, 山本昇平, 小野秀樹
    日本神経精神薬理学雑誌 33 81-83 2013年4月  
  • Shohei Yamamoto, Masahiro Ohsawa, Hideki Ono
    JOURNAL OF PHARMACOLOGICAL SCIENCES 121(1) 9-16 2013年1月  査読有り
    Neuropathic pain induces allodynia and hyperalgesia. In the spared nerve injury (SNI) model, marked mechanical hyperalgesia is manifested as prolongation of the duration of paw withdrawal after pin stimulation. We have previously reported that spinal ventral root discharges (after-discharges) after cessation of noxious mechanical stimulation applied to the corresponding hindpaw were prolonged in anesthetized spinalized rats. Since these after-discharges occurred through transient receptor potential (TRP) V1-positive fibers, these fibers could contribute to mechanical hyperalgesia. Therefore, we examined whether selective deletion of TRPV1-positive fibers by resiniferatoxin, an ultrapotent TRPV1 agonist, would affect the behavioral changes and ventral root discharges in SNI rats. Mechanical allodynia in the von Frey test, mechanical hyperalgesia after pin stimulation, and enhancement of ventral root discharges, but not thermal hyperalgesia in the plantar test, appeared in Wistar rats with SNI. Mechanical hyperalgesia was abolished by treatment with resiniferatoxin, whereas mechanical allodynia was not affected. Moreover, resiniferatoxin eliminated after-discharges completely. These results show that TRPV1-positive fibers do not participate in the mechanical allodynia caused by sensitization of A beta-fibers, but contribute to the enhancement of after-discharges and mechanical hyperalgesia following SNI. It is suggested that the mechanisms responsible for generating mechanical allodynia differ from those for prolongation of mechanical hyperalgesia.
  • Satoko Kimura, Yuka Niwa, Yui Iwajima, Yuko Nagano, Shohei Yamamoto, Yoshiaki Ohi, Yasuhiro Maeda, Yukihisa Kurono, Hideki Ono, Akira Haji
    BASIC & CLINICAL PHARMACOLOGY & TOXICOLOGY 111(4) 232-239 2012年10月  査読有り
    It has been reported that one of the serious adverse events after the treatment of oseltamivir phosphate (OP) for influenza patients is sudden death resulting from cardiorespiratory arrest. To investigate the aetiology of such an adverse consequence, we examined effects of OP (expressed as free base) on blood pressure and ventilation in anaesthetized rats with vagotomy. Intravenous OP (30200 mg/kg) caused dose-dependent hypotension and bradycardia in spontaneously breathing animals. Concomitantly with changes in blood pressure, the tracheal airflow increased. The ventilatory rate hastened during the injection and then transiently slowed around 1 min. after the administration (transient hypopnea). Thereafter, it gradually returned to control. The hypopnea increased with increasing dose and ventilatory arrest occurred at 200 mg/kg. Intraduodenal OP (5001000 mg/kg) provoked cardioventilatory arrest 72218 min. after the injection. Oseltamivir carboxylate (100200 mg/kg, i.v.), an active metabolite of OP, had no significant effect on ventilation and blood pressure. In artificially ventilated animals, intravenous OP caused slowing of the respiratory rate around 1 min. after the injection in a dose-dependent manner. This effect of OP waned in 5 min. after the administration. The amplitude of phrenic nerve discharge was not changed at lower doses (30100 mg/kg). The phrenic nerve stopped to discharge immediately after higher doses (150200 mg/kg). We demonstrated that OP causes central suppression of the respiratory function in rats and suggest a relationship between the OP-induced cardiorespiratory arrest and sudden death observed in influenza patients after taking OP.
  • S. Ohnami, A. Kato, K. Ogawa, S. Shinohara, H. Ono, M. Tanabe
    BRITISH JOURNAL OF PHARMACOLOGY 167(3) 537-547 2012年10月  査読有り
    BACKGROUND AND PURPOSE The analgesic action of 5-HT and noradrenaline reuptake inhibitors (SNRIs) on nociceptive synaptic transmission in the spinal cord is poorly understood. We investigated the effects of milnacipran, an SNRI, on C-fibre-evoked field potentials (FPs) in spinal long-term potentiation (LTP), a proposed synaptic mechanism of hypersensitivity, and on the FPs in a neuropathic pain model. EXPERIMENTAL APPROACH C-fibre-evoked FPs by electrical stimulation of the sciatic nerve fibres were recorded in the spinal dorsal horn of anaesthetized adult rats, and LTP was induced by high-frequency stimulation of the sciatic nerve fibres. A rat model of neuropathic pain was produced by L5 spinal nerve ligation and transection. KEY RESULTS Milnacipran produced prolonged inhibition of C-fibre-evoked FPs when applied spinally after the establishment of LTP of C-fibre-evoked FPs in naive animals. In the neuropathic pain model, spinal administration of milnacipran clearly reduced the basal C-fibre-evoked FPs. These inhibitory effects of milnacipran were blocked by spinal administration of methysergide, a 5-HT1/2 receptor antagonist, and yohimbine or idazoxan, a2-adrenoceptor antagonists. However, spinal administration of milnacipran in naive animals did not affect the basal C-fibre-evoked FPs and the induction of spinal LTP. CONCLUSION AND IMPLICATIONS Milnacipran inhibited C-fibre-mediated nociceptive synaptic transmission in the spinal dorsal horn after the establishment of spinal LTP and in the neuropathic pain model, by activating both spinal 5-hydroxytryptaminergic and noradrenergic systems. The condition-dependent inhibition of the C-fibre-mediated transmission by milnacipran could provide novel evidence regarding the analgesic mechanisms of SNRIs in chronic pain.
  • 大澤匡弘, 森直治, 川村和美, 島田雅彦, 二村昭彦, 山本昇平, 東口高志, 小野秀樹
    日本緩和医療薬学雑誌 5(2) 31-37 2012年6月  査読有り
  • Shohei Yamamoto, Mitsuo Tanabe, Hideki Ono
    JOURNAL OF PHARMACOLOGICAL SCIENCES 119(1) 82-90 2012年5月  査読有り
    Voltage-dependent Ca2+ channels (VDCCs) play a crucial role in the spinal pain transduction. We previously reported that nociceptive mechanical stimuli to the rat hindpaw evoked two types of ventral root discharges that increased during stimulation (during-discharges) and after cessation of stimulation (after-discharges). To explore the involvement of VDCCs in these ventral root discharges, several VDCC blockers were applied directly to the surface of the spinal cord. Spinalized rats were laminectomized. The fifth lumbar ventral root was sectioned and used for multi-unit efferent discharges recording. An agar pool was constructed on the first lumbar vertebra for drug application. Ethosuximide (a T-type VDCC blocker) had no effect on ventral root discharges. omega-Conotoxin GVIA (an N-type VDCC blocker) preferentially suppressed after-discharges. omega-Agatoxin IVA (a P/Q-type VDCC blocker), diltiazem, and verapamil (L-type VDCC blockers) nonselectively depressed both during- and after-discharges. The more selective L-type VDCC blocker nicardipine depressed only after-discharges and the depression was exhibited when nicardipine was microinjected into the dorsal horn, but not into the ventral horn. These findings suggested that N- and L-type VDCCs in the dorsal horn were involved in the generation of after-discharges and these blockers might be useful for treatment of persistent pain that involves the spinal pathway.
  • Masahiro Ohsawa, Junpei Mutoh, Shohei Yamamoto, Hideki Ono, Hiroaki Hisa
    JOURNAL OF PHARMACOLOGICAL SCIENCES 119(1) 102-106 2012年5月  査読有り
    Clinical and experimental observations indicated that 3-hydroxy-3-methylglutaryl CoA reductase inhibitor statins have pleiotropic effects. The present study determined the antinociceptive property of centrally administered simvastatin on the forrnalin-induced nociception in the mouse. Intrathecal administration of simvastatin at doses of 0.5 - 50 nmol dose-dependently attenuated the second, but not the first, phase of the formalin-induced nociception, which was partially reversed by mevalonate (5 mu mol). Intracerebroventricular injection of simvastatin (50 nmol) did not affect the formalin-induced nociception. These results suggest that simvastatin-induced antinociception is mediated by attenuation of the sensitization of spinal nociceptive transmission.
  • Masahiro Ohsawa, Junpei Mutoh, Megumi Asato, Shohei Yamamoto, Hideki Ono, Hiroaki Hisa, Junzo Kamei
    EUROPEAN JOURNAL OF PHARMACOLOGY 682(1-3) 56-61 2012年5月  査読有り
    Carnosine is a biologically active dipeptide that is found in fish and chicken muscle. Recent studies have revealed that carnosine has neuroprotective activity in zinc-induced neural cell apoptosis and ischemic stroke. In the present study, we examined the expression of carnosine in the spinal cord, and the antinociceptive potency of carnosine in a mouse model of inflammation-induced nociceptive pain. Immunohistochemical studies with antiserum against carnosine showed an abundance of carnosine-immunoreactivity in the dorsal horn of the mouse spinal cord. Double-immunostaining techniques revealed that carnosine was expressed in the neurons and astrocytes in the spinal cord. Oral administration of carnosine attenuated the number of writhing behaviors induced by the intraperitoneal administration of 0.6% acetic acid. Treatment with carnosine also attenuated the second phase, but not the first phase, of the nociceptive response to formalin. Moreover, intrathecal, but not intraplanter, administration of carnosine attenuated the second phase of the nociceptive response to formalin. Our immunohistochemical and behavioral data suggest that carnosine has antinociceptive effects toward inflammatory pain, which may be mediated by the attenuation of nociceptive sensitization in the spinal cord. (C) 2012 Elsevier B.V. All rights reserved.
  • 小野秀樹, 黒野幸久
    YAKUGAKU ZASSHI 132(1) 21-29 2012年1月  査読有り
    Eleven universities which have pharmacy, medical or nursing school, have cooperated in an attempt to build the human and material systems for 6-year pharmacy education and to apply them to practical pharmacy educations. Members are Nagoya City University, Gifu Pharmaceutical University, University of Shizuoka, Aichi Gakuin University, Kinjo Gakuin University, Meijo University, Suzuka University of Medical Science, Hamamatsu University School of Medicine, Mie University, Aichi Medical University and Fujita Health University. Tokai Cooperation Center for Clinical Pharmacy Education, the steering committee and 5 subcommittees established following projects; 1) WEB-based system for supplementary lesson of natural science (for freshmen), 2) FD (Faculty Development) activity (for teachers), 3) WEB-based data-base system of disease case for PBL (Problem-based Learning) and methods for practice of physical assessment (for 4th grade students), 4) WEB-based system for practical pharmacy training (for 5th grade students), 5) Matching and WEB system for graduation practice at university hospital (for 6th grade students).<br>
  • Daisuke Kodama, Hideki Ono, Mitsuo Tanabe
    PAIN 152(4) 809-817 2011年4月  査読有り
    Patients with chronic pain often have accompanying cognitive deficiency, which may reduce their quality of life and hamper efficient medical treatment. Alteration of extracellular glycine concentration may affect cognitive function and spinal pain signaling. In the present study, we assessed recognition memory by novel-object recognition and found that mice developing mechanical hypersensitivity after peripheral nerve injury exhibited impaired recognition ability for novelty, which was never observed in mice provided the selective glycine transporter 1 (GlyT1) inhibitor N-[3-(4'-fluorophenyl)-3-(4'-phenylphenoxy) propyl] sarcosine (NFPS) systemically. Although systemic NFPS generated analgesia via inhibitory effects of glycine in the spinal cord, the cognitive impairment in neuropathic mice was not restored upon relief of pain alone by intrathecal injection of NFPS. Whole-cell recordings were then made from hippocampal CA1 pyramidal neurons, and the effect of exogenously applied glycine or its endogenous increase by blockade of GlyT1 with NFPS on N-methyl-D-aspartate receptor-mediated excitatory postsynaptic currents (NMDA-EPSCs) was investigated in slices prepared from neuropathic mice and mice subjected to sham treatment. In slices from neuropathic mice, NMDA-EPSCs were less potentiated by glycine, whereas they were augmented by NFPS even at lower concentrations. After treating the slices with either NFPS or the glial-selective metabolic blocker fluoroacetate, glycine potentiated NMDA-EPSCs equally in slices from neuropathic and sham-treated mice. These findings imply that chronic pain has a crucial influence on hippocampal plasticity related to cognitive function, and strongly suggest that increasing the extracellular level of glycine via blockade of GlyT1 is a potential therapeutic approach for chronic pain with memory impairment. (C) 2010 International Association for the Study of Pain. Published by Elsevier B. V. All rights reserved.
  • Soichiro Ohnami, Mitsuo Tanabe, Shunji Shinohara, Keiko Takasu, Akira Kato, Hideki Ono
    PAIN 152(3) 623-631 2011年3月  査読有り
    Activity-dependent increases in the responsiveness of spinal neurons to their normal afferent input, termed central sensitization, have been suggested to play a key role in abnormal pain sensation. We investigated the role of distinct voltage-dependent calcium channel (VDCC) subtypes in the long-term potentiation (LTP) of C-fiber-evoked field potentials (FPs) recorded in the spinal dorsal horn of rats, that is, a synaptic model to describe central sensitization. When spinally applied, we observed that omega-conotoxin GVIA (omega-CgTx), an N-type VDCC antagonist, produced a dose-dependent and prolonged inhibition of basal C-fiber-evoked FPs in naive animals. omega-CgTx did not perturb the induction of LTP by high-frequency stimulation (HFS) of the sciatic nerve; however, potentiation was maintained at a lower level. Following the establishment of spinal LTP in naive animals, the inhibitory effect of omega-CgTx on C-fiber-evoked FPs was significantly increased. Furthermore, in animals with chronic pain produced via peripheral nerve injury, where spinal LTP was barely induced by HFS, basal C-fiber-evoked FPs were strongly inhibited by omega-CgTx. As a result, omega-CgTx exerted a similar inhibitory profile on C-fiber-evoked FPs following the establishment of spinal LTP and chronic pain. In contrast, spinally administered omega-agatoxin IVA (omega-Aga-IVA), a P/Q-type VDCC antagonist, showed little effect on C-fiber-evoked FPs either before or after the establishment of LTP, but strongly suppressed LTP induction. These results demonstrate the requirement of N- and P/Q-type VDCCs in the maintenance and induction of LTP in the spinal dorsal horn, respectively, and their distinct contribution to nociceptive synaptic transmission and its plasticity. (C) 2010 International Association for the Study of Pain. Published by Elsevier B. V. All rights reserved.
  • Keiko Takasu, Hideki Ono, Mitsuo Tanabe
    PAIN 151(1) 87-96 2010年10月  査読有り
    Hyperpolarization-activated cyclic nucleotide-gated cation channels (HCN channels) have large influences upon neuronal excitability. However, the participation of spinal HCN channels in chronic pain states, where pathological conditions are related to altered neuronal excitability, has not been clarified. Intraperitoneally (i.p.) or intrathecally (i.t.) administered ZD7288, a selective blocker of Ih channels, reduced thermal and mechanical hypersensitivity in mice under neuropathic conditions induced by partial ligation of the sciatic nerve, while no analgesic effect was observed in naive animals. Moreover, in the mouse formalin test, ZD7288 (i.p. and i.t.) reduced licking/biting behavior observed during the second phase without affecting the first phase. To further explore the pain-modulatory action of spinal HCN channels, whole-cell patch clamp recordings were made from visually identified substantia gelatinosa neurons in adult mouse spinal cord slices with an attached dorsal root, and A-fiber-and/or C-fiber-mediated monosynaptic excitatory postsynaptic currents (EPSCs) were evoked by electrical stimulation of the L4 or L5 dorsal root using a suction electrode. Bath-applied ZD7288 reduced A-fiber-and C-fiber-mediated monosynaptic EPSCs more preferentially in slices prepared from mice after peripheral nerve injury. In addition, ZD7288 reduced the frequency of miniature EPSCs without affecting their amplitude in cells receiving monosynaptic afferent inputs, indicating that it inhibits EPSCs via presynaptic mechanisms. The present behavioral and electrophysiological data suggest that spinal HCN channels, most likely at primary afferent terminals, contribute to the maintenance of chronic pain. (C) 2010 International Association for the Study of Pain. Published by Elsevier B.V. All rights reserved.
  • Mitsuo Tanabe, Azusa Nitta, Hideki Ono
    JOURNAL OF PHARMACOLOGICAL SCIENCES 113(4) 378-386 2010年8月  査読有り
    Recent evidence indicates that strychnine-sensitive glycine receptors are located in upper brain regions including the hippocampus. Because of excitatory effects of glycine via facilitation of NMDA-receptor function, however, the net effects of increased extracellular glycine on neuronal excitability in either physiological or pathophysiological conditions are mostly unclear. Here, we addressed the potential neuroprotective effect of either exogenous application of glycine and taurine, which are both strychnine-sensitive glycine-receptor agonists, or an endogenous increase of glycine via blockade of glycine transporter 1 (GlyT1) by assessing their ability to facilitate the functional recovery of field excitatory postsynaptic potentials (fEPSPs) after termination of brief oxygen/glucose deprivation (OGD) in the CA1 region in mouse hippocampal slices. Glycine and taurine promoted restoration of the fEPSPs after reperfusion, but this was never observed in the presence of strychnine. Interestingly, glycine and taurine appeared to generate neuroprotective effects only at their optimum concentration range. By contrast, blockade of GlyT1 by N-[3-(4&apos;-fluorophenyl)-3-(4&apos;-phenylphenoxy)propyl]sarcosine or sarcosine did not elicit significant neuroprotection. These results suggest that activation of strychnine-sensitive glycine receptors potentially produces neuroprotection against metabolic stress such as OGD. However, GlyT1 inhibition is unlikely to elicit a sufficient increase in the extracellular level of glycine to generate neuroprotection.
  • Tanabe M, Takasu K, Ono H
    Nihon yakurigaku zasshi. Folia pharmacologica Japonica 134(6) 299-303 2009年12月  査読有り
    抗てんかん薬ガバペンチンは,欧米において神経因性疼痛治療薬としての地位を確立しているが,その作用メカニズムについては未解明な部分が多い.我々はその作用部位として上位中枢に焦点を当てた研究を行い,脳室内投与したガバペンチンが神経損傷(マウス坐骨神経部分結紮モデル)後の疼痛症状(熱痛覚過敏および機械アロディニア)に対し障害依存的な鎮痛作用を発揮することを示し,ガバペンチン全身投与後の鎮痛作用において,上位中枢を介する効果が大きく寄与することを見出した.ガバペンチンの全身投与あるいは脳室内投与によって引き起こされる鎮痛効果は,脳幹から脊髄へ下行するノルアドレナリン(NA)神経を消失させると大幅に減弱し,また,α2-アドレナリン受容体アンタゴニストヨヒンビンの全身投与や脊髄内投与によって同様に減弱した.脳室内投与したガバペンチンが脊髄腰部膨大部のNA代謝回転を神経障害依存的に促進させたことからも,上位中枢に作用したガバペンチンが下行性NA神経を介して脊髄内においてNA遊離を増加させ,α2-アドレナリン受容体を介した鎮痛効果を発揮すると考えられる.さらに,坐骨神経部分結紮による神経障害後に作製したマウス脳幹スライスの青斑核ニューロンにおいて,ガバペンチンはGABA性の抑制性シナプス伝達をシナプス前性に抑制することを明らかにした.Sham手術マウス由来のスライスではガバペンチンはこの抑制性シナプス伝達抑制作用を示さず,また,神経障害後でも興奮性シナプス伝達に対しては影響を及ぼさなかった.これらの研究結果より,ガバペンチンは青斑核においてGABA性の抑制性入力を抑制することによって青斑核ニューロンを脱抑制し,下行性NA疼痛抑制経路を活性化させて神経因性疼痛を緩解することが示唆された.<br>
  • Keiko Takasu, Yu Kinoshita, Hideki Ono, Mitsuo Tanabe
    JOURNAL OF PHARMACOLOGICAL SCIENCES 110(2) 223-226 2009年6月  査読有り
    We have recently shown that gabapentin generates protein kinase A (PKA)-dependent presynaptic inhibition of GABAergic synaptic transmission in locus coeruleus (LC) neurons only under neuropathic states. To verify behaviorally this in vitro electrophysiological finding, the PKA inhibitor H-89 was injected intracerebroventricularly (i.c.v.) before supraspinal application of gabapentin in mice developing thermal and mechanical hypersensitivity after peripheral nerve injury. H-89 dose-dependently attenuated the analgesic effects of i.c.v.-injected gabapentin, Suggesting that PKA-dependent removal of GABAergic inhibition of LC neurons is the most plausible synaptic mechanism underlying the supraspinally mediated analgesic effects of gabapentin involving activation of the descending noradrenergic pain-inhibitory system.
  • Mitsuo Tanabe, Koto Ono, Motoko Honda, Hideki Ono
    EUROPEAN JOURNAL OF PHARMACOLOGY 609(1-3) 65-68 2009年5月  査読有り
    The antiepileptic drugs gabapentin and pregabalin exhibit well-established analgesic effects in patients with several neuropathic conditions. In the present study, we examined their effects on mechanical hypersensitivity in mice subjected to weight-drop spinal cord injury. Hindlimb motor function and mechanical hypersensitivity were evaluated using the Basso-Beattie-Bresnahan (BBB) locomotor rating scale and the von Frey test, respectively, for 4 weeks after spinal cord injury. Despite gradual recovery of hindlimb motor function after spinal cord injury, mice exhibited continuous development of mechanical hypersensitivity. Gabapentin (30 and 100 mg/kg) and pregabalin (10 and 30 mg/kg), administered intraperitoneally on the 28th day after spinal cord injury, reduced mechanical hypersensitivity in a dose-dependent manner. These results suggest that gabapentin and pregabalin could be useful therapeutic tools for patients with neuropathic pain after spinal cord injury. (C) 2009 Elsevier B.V. All rights reserved.
  • Tanabe M, Nakano T, Honda M, Ono H
    Journal of pharmacological sciences 109(3) 444-448 2009年3月  査読有り
  • Mitsuo Tanabe, Yoshinori Nagatani, Kazuya Saitoh, Keiko Takasu, Hideki Ono
    NEUROPHARMACOLOGY 56(3) 702-708 2009年3月  査読有り
    Nitric oxide synthase (NOS) isoforms and NO downstream signal pathways involved spinally in the maintenance of thermal and mechanical hypersensitivity were assessed in a mouse model of neuropathic pain developing after partial ligation of the sciatic nerve. Intrathecal injection of the NOS inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME), the highly selective neuronal NOS (nNOS) inhibitor N(omega)-propyl-L-arginine and the potent selective inducible NOS (iNOS) inhibitor 2-amino-5,6-dihydro-6-methyl-4H-1,3-thiazine hydrochloride (AMT) exerted dose-dependent analgesic effects on thermal and mechanical hypersensitivity, which were assessed by the plantar and von Frey tests, respectively, suggesting that both nNOS and iNOS participate in producing NO to maintain neuropathic pain. Since the selective inhibitor of NO-sensitive guanylyl cyclase 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) and the guanosine 3&apos;,5&apos;-cyclic monophosphate (cGMP)-dependent protein kinase (PKG) inhibitor Rp-8-pCPT-cGMPS intrathecally exerted dose-dependent analgesic effects on thermal and mechanical hypersensitivity. spinally released NO most likely stimulates the NO-cGMP-PKG pathway. Moreover, the superoxide dismutase mimetic 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl (TEMPOL), a potent superoxide scavenger, reduced the.-Mal and mechanical hypersensitivity when administered intrathecally, suggesting that spinal release of superoxide, which can then react with NO to produce peroxynitrite, also appears to mediate neuropathic pain. Finally, intrathecal injection of phenyl-N-tert-butylnitrone (PBN), a reactive oxygen species (ROS) scavenger, ameliorated thermal and mechanical hypersensitivity, thus further confirming the importance of ROS including NO and superoxide in the maintenance of neuropathic pain. Together, the present results demonstrate that NO, produced presumably via nNOS and iNOS in the spinal cord, mediates the maintenance of neuropathic pain following peripheral nerve injury through both the NO-cGMP-PKG and the NO-peroxynitrite pathways. (C) 2008 Elsevier Ltd. All rights reserved.
  • 田辺光男, 高須景子, 小野秀樹
    日本薬理学雑誌 134(6) 299-303-303 2009年  
    抗てんかん薬ガバペンチンは,欧米において神経因性疼痛治療薬としての地位を確立しているが,その作用メカニズムについては未解明な部分が多い.我々はその作用部位として上位中枢に焦点を当てた研究を行い,脳室内投与したガバペンチンが神経損傷(マウス坐骨神経部分結紮モデル)後の疼痛症状(熱痛覚過敏および機械アロディニア)に対し障害依存的な鎮痛作用を発揮することを示し,ガバペンチン全身投与後の鎮痛作用において,上位中枢を介する効果が大きく寄与することを見出した.ガバペンチンの全身投与あるいは脳室内投与によって引き起こされる鎮痛効果は,脳幹から脊髄へ下行するノルアドレナリン(NA)神経を消失させると大幅に減弱し,また,α2-アドレナリン受容体アンタゴニストヨヒンビンの全身投与や脊髄内投与によって同様に減弱した.脳室内投与したガバペンチンが脊髄腰部膨大部のNA代謝回転を神経障害依存的に促進させたことからも,上位中枢に作用したガバペンチンが下行性NA神経を介して脊髄内においてNA遊離を増加させ,α2-アドレナリン受容体を介した鎮痛効果を発揮すると考えられる.さらに,坐骨神経部分結紮による神経障害後に作製したマウス脳幹スライスの青斑核ニューロンにおいて,ガバペンチンはGABA性の抑制性シナプス伝達をシナプス前性に抑制することを明らかにした.Sham手術マウス由来のスライスではガバペンチンはこの抑制性シナプス伝達抑制作用を示さず,また,神経障害後でも興奮性シナプス伝達に対しては影響を及ぼさなかった.これらの研究結果より,ガバペンチンは青斑核においてGABA性の抑制性入力を抑制することによって青斑核ニューロンを脱抑制し,下行性NA疼痛抑制経路を活性化させて神経因性疼痛を緩解することが示唆された.<br>
  • Mitsuo Tanabe, Keiko Takasu, Yuichi Takeuchi, Hideki Ono
    JOURNAL OF NEUROSCIENCE RESEARCH 86(15) 3258-3264 2008年11月  査読有り
    The anti hypersensitivity actions of gabapentin and pregabalin have been well characterized in a large number of studies, although the underlying mechanisms have yet to be defined. We have been focusing on the supraspinal structure as a possible site for their action and have demonstrated that intracerebroventricular (i.c.v.) administration of gabapentin and pregabalin indeed decreases thermal and mechanical hypersensitivity in a murine chronic pain model involving partial ligation of the sciatic nerve. This novel supraspinally mediated analgesic effect was markedly suppressed by either depletion of central noradrenaline (NA) or blockade of spinal alpha(2)-adrenergic receptors. Moreover, i.c.v. injection of gabapentin and pregabalin increased spinal NA turnover in mice only after peripheral nerve injury. In locus coeruleus (LC) neurons in brainstem slices prepared from mice after peripheral nerve injury, gabapentin reduced the gamma-aminobutyric acid (GABA) type A receptor-mediated inhibitory postsynaptic currents (IPSCs). Glutamate-mediated excitatory synaptic transmission was hardly affected. Moreover, gabapentin did not reduce IPSCs in slices taken from mice given a sham operation. Although gabapentin altered neither the amplitude nor the frequency of miniature IPSCs, it reduced IPSCs together with an increase in the paired-pulse ratio, suggesting that gabapentin acts on the presynaptic GABAergic nerve terminals in the LC. Together, the data suggest that gabapentin presynaptically reduces GABAergic synaptic transmission, thereby removing the inhibitory influence on LC neurons only in neuropathic pain states, leading to activation of the descending noradrenergic system. (C) 2008 Wiley-Liss, Inc.
  • Tanabe M, Hashimoto M, Ono H
    European journal of pharmacology 589(1-3) 102-105 2008年7月  査読有り
  • Mitsuo Tanabe, Takuhiro Murakami, Hideki Ono
    JOURNAL OF PHARMACOLOGICAL SCIENCES 107(2) 213-220 2008年6月  査読有り
    We evaluated the effects of zonisamide on inflammatory and neuropathic pain using the mouse formalin test and streptozotocin (STZ)-induced diabetic mice with a reduced withdrawal threshold to mechanical stimuli, respectively. When administered systemically (subcutaneously, s.c.), intracerebroventricularly (i.c.v.) or intrathecally (i.t.) before formalin injection, zonisamide (3 and 10 mg/kg, s.c., 10 and 30,mu g, i.c.v., or i.t.) significantly reduced licking/biting behavior during the second phase of the formalin test in a dose-dependent manner. However, zonisamide (30,mu g, i.t.) did not affect the second phase of the formalin test when given after the first phase, suggesting that it can prevent the development of injury-induced hyperexcitability of the spinal dorsal horn triggered by the repetitive nociceptive input during the first phase. Moreover, zonisamide administered into the dorsal hindpaw ipsilateral but not contralateral to the formalin injection partly reduced the second phase. Thus it is likely that zonisamide generates analgesic effects in the formalin test via both central and peripheral mechanisms. In mice with STZ-induced diabetes, zonisamide (10 and 30 mg/kg, s.c. or 10 and 30 mu g, i.t.) reversed the mechanical hyperexcitability. Our results suggest that zonisamide can be a useful therapeutic agent, presumably for both prevention and reversal of pathophysiologic pain.
  • Mitsuo Tanabe, Keiko Takasu, Sachiko Yamaguchi, Daisuke Kodama, Hideki Ono
    ANESTHESIOLOGY 108(5) 929-937 2008年5月  査読有り
    Background: Impaired excitatory and inhibitory balance in the spinal dorsal horn has a crucial role in the pathophysiology of chronic pain. The authors addressed the therapeutic impact of increasing spinal glycine applied exogenously or via blockade of glycine transporter 1 using its selective inhibitors sarcosine and N-[3-(4'-fluorophenyl)-3-(4'-phenylphenoxy)propyl]sarcosine on neuropathic and inflammatory pain in mice. Methods: Mice with thermal and mechanical hypersensitivity after partial ligation of the sciatic nerve (Seltzer model) or mice with mechanical hypersensitivity after streptozotocin injection received intrathecal injection of glycine, sarcosine, and N-[3-(4'-fluorophenyl)-3-(4'-phenylphenoxy)propyl]sarcosine. These drugs were also intrathecally injected in mice to assess their effects on formalin-evoked nociceptive behaviors. The supraspinal effect of blockade of glycine transporter 1 was studied on tetanus-induced long-term potentiation of the Schaffer-collateral synapses in hippocampal slices prepared from Seltzer model mice. Results: Glycine, sarcosine, and N-[3-(4'-fluorophenyl)-3-(4'-phenylphenoxy)propyl]sarcosine ameliorated thermal and mechanical hypersensitivity in Seltzer model mice, and reduced mechanical hypersensitivity in streptozotocin-injected diabetic mice. Moreover, they selectively inhibited the second phase of formalin-evoked licking/biting behavior. In hippocampal slices prepared from Seltzer model mice, long-term potentiation was maintained at a significantly lower level than that in sham-treated mice. Such impairment of long-term potentiation was never observed when it was induced in the presence of N-[3-(4'-fluorophenyl)-3-(4'-phenylphenoxy)propyl]sarcosine. Conclusions: An increase in endogenous glycine via glycine transporter 1 blockade not only results in a net inhibitory influence on pain transmission at the spinal level but also supraspinally relieves decreased synaptic efficacy presumably related to cognitive disturbance often described in patients with chronic pain.
  • Keiko Takasu, Hideki Ono, Mitsuo Tanabe
    JOURNAL OF NEUROCHEMISTRY 105(3) 933-942 2008年5月  査読有り
    We have previously demonstrated that gabapentin supraspinally activates the descending noradrenergic system to ameliorate pain hypersensitivity in mice with partial nerve ligation. To clarify the supraspinal mechanism of action of gabapentin, whole-cell patch-clamp recordings were performed on locus coeruleus (LC) neurons in brainstem slices prepared from mice after peripheral nerve injury or mice subjected to a sham-operation, and the effects of gabapentin in the modulation of synaptic transmission were studied. Bath application of gabapentin (10, 30 and 100 mu M) in a concentration-dependent manner reduced the GABA(A) receptor-mediated inhibitory post-synaptic currents (IPSCs) in slices prepared from partially nerve-ligated mice, whereas glutamate-mediated excitatory post-synaptic currents were hardly affected. By contrast, gabapentin did not reduce IPSCs in slices taken from mice given a sham operation. Although gabapentin altered neither the amplitude nor the frequency of miniature IPSCs, it reduced IPSCs together with an increase in the paired-pulse ratio, suggesting that gabapentin acts on the pre-synaptic GABAergic nerve terminals in the LC. As the protein kinase A (PKA) inhibitor H-89 but not the protein kinase C inhibitor chelerythrine abolished the inhibitory action of gabapentin on IPSCs, PKA-mediated phosphorylation seems to be important for supraspinal gabapentin responsiveness in neuropathic conditions. Together, gabapentin generates PKA-dependent pre-synaptic inhibition of GABAergic synaptic transmission, and thereby removes the inhibitory influence on LC neurons only under neuropathic pain states. These findings provide crucial evidence of how supraspinally acting gabapentin recruits the descending noradrenergic system.
  • Hideki Ono, Yuko Nagano, Noriaki Matsunami, Shinichi Sugiyama, Shohei Yamamoto, Mitsuo Tanabe
    BIOLOGICAL & PHARMACEUTICAL BULLETIN 31(4) 638-642 2008年4月  査読有り
    Oseltamivir phosphate (Tamiflu), an anti-influenza virus drug, is hydrolyzed by carboxylesterase to an active metabolite. The metabolite inhibits the influenza virus-specific neuraminidase. In this study, the effects of oseltamivir on normal core body temperature were studied in mice. Oseltamivir (30-300 mg/kg, intraperitoneally (i.p.) and 100-1000 mg/kg, orally (p.o.)) dose-dependently lowered the body temperature. The effects of oseltamivir (p.o.) continued longer than those of oseltamivir (i.p.), and approximately triple doses of oral oseltamivir were needed to produce the same peak effects as intraperitoneal oseltamivir. The non-steroidal anti-inflammatory drug diclofenac (1-30 mg/kg, i.p.) did not affect body temperature, and (at 30 and 60 mg/kg, s.c.) did not interact with the hypothermic effects of oseltamivir (100 mg/kg, i.p.). Zanamivir, which also inhibits neuraminidase, did not produce hypothermia at doses of 100 and 300 mg/kg, i.p. Clopidogrel (100, 300 mg/kg, i.p.), which is metabolized by the same carboxylesterase, tended to decrease the hypothermic effects of oseltamivir (100 mg/kg, i.p.). These results suggest that the hypothermic effects of oseltamivir are due to its hydrolytic metabolite, and that the hypothermia observed in mice has some relationship to the antipyretic effects and severe hypothermia (adverse event) observed in influenza patients after taking oseltamivir.
  • Yuichi Takeuchi, Keiko Takasu, Hideki Ono, Mitsuo Tanabe
    NEUROPHARMACOLOGY 53(7) 842-853 2007年12月  査読有り
    We have previously demonstrated that gabapentin supraspinally activates the descending noradrenergic system to alleviate neuropathic pain. In this study, we investigated whether pregabalin, an antiepileptic and analgesic drug that is also designed as a structural analogue of gamma-aminobutyric acid (GABA), exhibits supraspinal analgesic effects similar to those of gabapentin involving the descending noradrenergic system. Both systemically (intraperitoneally; i.p.) and locally (intracerebroventricularly or intrathecally; i.c.v. or i.t.) injected pregabalin reduced thermal and mechanical hypersensitivity in a murine chronic pain model that was prepared by partial ligation of the sciatic nerve (the Seltzer model), suggesting that pregabalin acts at both supraspinal and spinal loci. The supraspinal analgesic action of pregabalin was observed only after peripheral nerve injury. and pregabalin (i.p. and i.c.v.) did not affect acute thermal and mechanical nociception, Depletion of spinal noradrenaline (NA) or pharmacological blockade of spinal alpha(2)-adrenoceptors with yohimbine i.p. or i.t.), but not alpha(1)-adrenoceptors with prazosin (i.p.), reduced the analgesic effects of pregabalin (i.p. or i.c.v.) on thermal and mechanical hypersensitivity. Moreover. i,c.v.-administered pregabalin dose-dependently increased the spinal 4-hydroxy-3-methoxyphenylglycol (MHPG) content and the MHPG/NA ratio only in mice with neuropathic pain, whereas the concentrations of NA, serotonin, 5-hydroxyindoleacetic acid and dopamine were unchanged, demonstrating that supraspinal pregabalin accelerated the spinal turnover of NA. Together, these results indicate that pregabalin Supraspinally activates the descending noradrenergic pain inhibitory system coupled with spinal alpha(2)-adrenoceptors to ameliorate neuropathic pain. (c) 2007 Elsevier Ltd. All rights reserved.
  • Daisuke Kodama, Hideki Ono, Mitsuo Tanabe
    EUROPEAN JOURNAL OF PHARMACOLOGY 574(2-3) 127-132 2007年11月  査読有り
    It has been clinically reported that patients with chronic pain often have accompanying cognitive deficiency, which hampers efficient medical treatment. In the present study, we investigated whether hippocampal synaptic plasticity, which has been considered to be a cellular model of learning and memory, could be influenced by chronic pain conditions using a murine model of neuropathic pain prepared by partial ligation of the sciatic nerve (the Seltzer model). In slices obtained from neuropathic animals, tetanus-induced long-term potentiation of CA1 hippocampal synaptic transmission was impaired, whereas long-term depression induced by low-frequency stimulation was similar in neuropathic and shamtreated (control) animals. Bath application of the beta-adrenoceptor agonist isoproterenol or the beta-adrenoceptor antagonist propranolol diminished the difference of synaptic plasticity between neuropathic and control mice. In the presence of isoproterenol, long-term potentiation was successfully induced in neuropathic mice. By contrast, long-term potentiation in sham-treated mice was impaired by propranolol which did not alter the already impaired long-term potentiation after peripheral nerve injury. These results suggest that beta-adrenergic functions are changed in chronic pain conditions, which may underlie the deficiency of long-term potentiation. (c) 2007 Elsevier B.V All rights reserved.
  • Mitsuo Tanabe, Yuichi Takeuchi, Hideki Ono
    JOURNAL OF PHARMACOLOGICAL SCIENCES 104(4) 335-340 2007年8月  査読有り
    We have previously demonstrated that the antiepileptic drug zonisamide supraspinally generates analgesic effects on thermal and mechanical hypersensitivity in mice after peripheral nerve injury. To further establish the neurochemical basis for the supraspinally mediated analgesic action of zonisamide, we measured spinal noradrenaline (NA), 3-methoxy-4hydroxyphenyleneglycol (MHPG), serotonin (5-HT), 5-hydroxyindoleacetic acid (5-HIAA), and dopamine (DA) contents using HPLC with electrochemical detection in a murine neuropathic pain model that was prepared by partial ligation of the sciatic nerve (Seltzer model). Intraperitoneally or intracerebroventricularly administered zonisamide (50 mg/kg, i.p. and 30 mu g, i.c.v., respectively), which almost completely reduced mechanical hypersensitivity, did not elicit any changes in spinal NA, MHPG, 5-HT, 5-HIAA, and DA contents. Moreover, the effectiveness of i.p. or i.c.v. administered zonisamide at reducing thermal and mechanical hypersensitivity was not influenced by intrathecally administered yohimbine (3,mu g), an alpha(2)-adrenergic receptor antagonist. Thus, it appears that the supraspinally mediated analgesic effects of zonisamide are independent of the descending monoaminergic pain inhibitory system.
  • M. Tanabe, Y. Tokuda, K. Takasu, K. Ono, M. Honda, H. Ono
    BRITISH JOURNAL OF PHARMACOLOGY 150(4) 403-414 2007年2月  査読有り
    Background and purpose: Exogenously administered thyrotropin-releasing hormone (TRH) is known to exert potent but short-acting centrally-mediated antinociceptive effects. We sought to investigate the mechanisms underlying these effects using the synthetic TRH analogue taltirelin, focusing on the descending monoaminergic systems in mice. Experimental approach: The mice received systemic or local injections of taltirelin combined with either central noradrenaline (NA) or 5-hydroxytryptamine (5-HT) depletion by 6-hydroxydopamine (6-OHDA) or DL-p-chlorophenylalanine (PCPA), respectively, or blockade of their receptors. The degree of antinociception was determined using the tail flick and tail pressure tests. Key results: Subcutaneously (s.c.) administered taltirelin exhibited dose-dependent antinociceptive effects in the tail flick and tail pressure tests. These effects appeared to be primarily supraspinally mediated, since intracerebroventricularly (i.c.v.) but not intrathecally (i.t.) injected taltirelin generated similar effects. Depletion of central NA abolished only the analgesic effect of taltirelin (s.c. and i.c.v.) on mechanical nociception. By contrast, depletion of central 5-HT abolished only its analgesic effect on thermal nociception. Intraperitoneal (i.p.) and i.t. injection of the alpha(2)-adrenoceptor antagonist yohimbine respectively reduced the analgesic effect of taltirelin (s.c. and i.c.v.) on mechanical nociception. By contrast, the 5-HT1A receptor antagonist WAY-100635 (i.p. and i.t.) reduced the effect of taltirelin (s.c. and i.c.v.) on thermal nociception. Neither the 5-HT2 receptor antagonist ketanserin nor the opioid receptor antagonist naloxone altered the antinociceptive effect of taltirelin. Conclusions and Implications: These findings suggest that taltirelin activates the descending noradrenergic and serotonergic pain inhibitory systems, respectively, to exert its analgesic effects on mechanical and thermal nociception.
  • Yuichi Takeuchi, Keiko Takasu, Motoko Honda, Hideki Ono, Mitsuo Tanabe
    EUROPEAN JOURNAL OF PHARMACOLOGY 556(1-3) 69-74 2007年2月  査読有り
    We have previously demonstrated that gabapentin supraspinally activates the descending noradrenergic system to produce analgesic effects after peripheral nerve injury. To further establish the neurochemical basis for its supraspinally mediated analgesic action, concentrations of spinal noradrenaline, 4-hydroxy-3-methoxyphenylglycol (MHPG), serotonin (5-HT), 5-hydroxyindoleacetic acid (5-HIAA) and dopamine were measured using high-performance liquid chromatography in a murine neuropathic pain model that was prepared by partial ligation of the sciatic nerve (the Seltzer model). Intracerebroventricularly (i.c.v.) administered gabapentin (100 and 300 mu g) increased the spinal MHPG concentration and the MHPG/noradrenaline ratio and alleviated mechanical hypersensitivity, whereas the concentrations of noradrenaline, 5-HT, 5-HIAA and dopamine were unchanged. By contrast, i.c.v. gabapentin neither affected the spinal MHPG concentration and MHPG/noradrenaline ratio nor exhibited analgesic effects in animals subjected to a sham operation. In addition, spinal monoamine levels in ligated animals were not changed after intrathecal administration of gabapentin which however generated analgesic effects. Thus, the supraspinally mediated analgesic effects of gabapentin are correlated with an increase in spinal noradrenaline turnover. (c) 2006 Elsevier B.V. All rights reserved.
  • Mitsuo Tanabe, Motoki Umeda, Motoko Honda, Hideki Ono
    EUROPEAN JOURNAL OF PHARMACOLOGY 553(1-3) 104-108 2006年12月  査読有り
    Antiepileptic drugs have been shown to reduce the severity of neurodegeneration resulting from stroke or brain injury. In the present study, we evaluated the effects of the antiepileptic drugs phenytoin and carbamazepine on the time course of changes in the population spike (PS) during brief oxygen/glucose deprivation (OGD) in the CA1 pyramidal region of rat hippocampal slices in vitro. After introducing simulated ischemia by OGD, the PS was initially inhibited, followed by transient recovery and subsequent reinhibition again concomitantly with disappearance of the presynaptic volley (PV). The slices were then reperfused with oxygen/glucose-containing solution. Both phenytoin and carbamazepine (30 and 100 mu M each) concentration-dependently delayed the initial inhibition and the time to transient recovery of the PS during OGD, thus prolonging the time until disappearance of the PV. However, they significantly promoted restoration of the PS after reperfusion. These results suggest that treatment with phenytoin and carbamazepine increases the resistance of tissue to energy deprivation, as evidenced by the facilitated post-ischemic recovery of the PS, despite prolonged ischemia. (c) 2006 Elsevier B.V. All rights reserved.
  • Mitsuo Tanabe, Hiroyuki Murakami, Motoko Honda, Hideki Ono
    EXPERIMENTAL NEUROLOGY 202(2) 280-286 2006年12月  査読有り
    C-fiber-evoked field potentials in response to electrical stimulation of the sciatic nerve were recorded in the dorsal horn of the rat lumbar spinal cord, and their long-term potentiation (LTP) was induced by high-frequency stimulation applied on the sciatic nerve as a synaptic model of hypersensitivity underlying an increased efficacy of nociceptive transmission. We evaluated the effect of gabapentin on the basal C-fiber-evoked field potentials and their established LTP. Intravenously administered gabapentin (10 and 30 mg/kg, i.v.) reduced the LTP of C-fiber-evoked field potentials in a dose-dependent manner when applied 60 min after establishment of the LTP. However, gabapentin did not affect the basal C-fiber-evoked field potentials or induction of the LTP. Thus, gabapentin was effective only in sensitized conditions. By contrast, morphine HCl (1 and 3 or 10 mg/kg, i.v.) reduced both the basal responses and their established LTP. The combination of gabapentin and morphine at lower doses of each drug appeared to result in a stronger reduction on the established LTP than that of each drug alone, suggesting that combination therapy can generate better analgesia in the treatment of chronic pain. (c) 2006 Elsevier Inc. All rights reserved.
  • Motoko Honda, Mitsuo Tanabe, Hideki Ono
    EXPERIMENTAL NEUROLOGY 202(1) 258-261 2006年11月  査読有り
    We studied changes in the spinal segmental reflex and serotonergic (5-HT) responses in rats after spinal cord injury (SCI) produced by the weight-dropping method at the T8 level. The spinal monosynaptic reflex amplitude (MSR) was recorded from the L5 ventral root following stimulation of the ipsilateral L5 dorsal root. The 5-HT precursor L-5-hydroxytryptophan (L-5-HTP) depressed NISR in the spinal cord injured rats but not in normal rats. We investigated whether the SCI-specific depression of NISR by L-5-HTP was attributable to postsynaptic supersensitivity of 5-HT receptors or presynaptic loss of the 5-HT uptake system. Sumatriptan, a selective 5-HT1B/1D receptor agonist that is not taken up by 5-HT transporters, depressed the MSR similarly in both SCI and normal rats, suggesting that SCI resulted in the loss of 5-HT terminals and not postsynaptic supersensitivity of 5-HT receptors. (c) 2006 Elsevier Inc. All rights reserved.
  • Motoko Honda, Kunitoshi Uchida, Mitsuo Tanabe, Hideki Ono
    NEUROPHARMACOLOGY 51(4) 866-872 2006年9月  査読有り
    There is an association between depression and chronic pain, and some antidepressants exert antinociceptive effects in humans and laboratory animals. We examined the effects of fluvoxamine, a selective serotonin reuptake inhibitor, on mechanical allodynia and its mechanism of action in the mouse chronic pain model, which was prepared by partially ligating the sciatic nerve. The antiallodynic effect was measured using the von Frey test. Fluvoxamine produced antiallodynic effects following both systemic and intrathecal administration. In 5-hydroxytryptamine (5-HT)-depleted mice, prepared by intracerebroventricular injection of 5,7-dihyroxytryptamine, the fluvoxamine-induced antiallodynic effect was significantly attenuated. The antiallodynic effects of systemic fluvoxamine were also reduced by both systemic and intrathecal administration of ketanserin, a 5-HT2A/2C receptor antagonist. In addition, fluvoxamine also induced antinociceptive effect in the acute paw pressure test, and this effect was antagonized by the 5-HT3 receptor antagonist granisetron. These results indicate that fluvoxamine exerts its antiallodynic effects on neuropathic pain via descending 5-HT fibers and spinal 5-HT2A or 5-HT2C receptors, and the antinociception on acute mechanical pain via 5-HT3 receptors. (c) 2006 Elsevier Ltd. All rights reserved.
  • K Takasu, M Honda, H Ono, M Tanabe
    BRITISH JOURNAL OF PHARMACOLOGY 148(2) 233-244 2006年5月  査読有り
    1 After partial nerve injury, the central analgesic effect of systemically administered gabapentin is mediated by both supraspinal and spinal actions. We further evaluate the mechanisms related to the supraspinally mediated analgesic actions of gabapentin involving the descending noradrenergic system. 2 Intracerebroventricularly (i.c.v.) administered gabapentin (100 mu g) decreased thermal and mechanical hypersensitivity in a murine chronic pain model that was prepared by partial ligation of the sciatic nerve. These effects were abolished by intrathecal (i.t.) injection of either yohimbine (3 mu g) or idazoxan (3 mu g), alpha 2-adrenergic receptor antagonists. 3 Pretreatment with atropine (0.3 mg kg(-1), i.p. or 0.1 mu g, i.t.), a muscarinic receptor antagonist, completely suppressed the effect of i.c.v.-injected gabapentin on mechanical hypersensitivity, whereas its effect on thermal hypersensitivity remained unchanged. Similar effects were obtained with pirenzepine (0.1 mg, i.t.), a selective M-1-muscarinic receptor antagonist, but not with methoctramine (0.1 and 0.3 mg, i.t.), a selective M-2-muscarinic receptor antagonist. 4 The cholinesterase inhibitor neostigmine (0.3 ng, i.t.) potentiated only the analgesic effect of i. c. v. gabapentin on mechanical hypersensitivity, confirming spinal acetylcholine release downstream of the supraspinal action of gabapentin. 5 Moreover, the effect of i. c. v. gabapentin on mechanical but not thermal hypersensitivity was reduced by i.t. injection of L-NAME (3 mg) or L-NMMA (10 mg), both of which are nitric oxide (NO) synthase inhibitors. 6 Systemically administered naloxone (10 mg kg(-1), i. p.), an opioid receptor antagonist, failed to suppress the analgesic actions of i. c. v. gabapentin, indicating that opioid receptors are not involved in activation of the descending noradrenergic system by gabapentin. 7 Thus, the supraspinally mediated effect of gabapentin on mechanical hypersensitivity involves activation of spinal alpha(2)-adrenergic receptors followed by muscarinic receptors ( most likely M-1) and the NO cascade. In contrast, the effect of supraspinal gabapentin on thermal hypersensitivity is independent of the spinal cholinergic-NO system.
  • S Yamamoto, M Honda, M Tanabe, H Ono
    BRAIN RESEARCH 1082(1) 115-123 2006年4月  査読有り
    Nociceptive signals are transmitted to the spinal dorsal horn via primary afferent fibers, and the signals induce withdrawal reflexes by activating spinal motoneurons in the ventral horn. Therefore, nociceptive stimuli increase motoneuronal firing and ventral root discharges This study was aimed to develop a method for the study of pain mechanisms and analgesics by recording ventral root discharges. Spinalized rats were laminectomized in the lumbo-sacral region. The fifth lumbar ventral root was sectioned and placed on a pair of wire electrodes Multi unit efferent discharges from the ventral root were increased by mechanical stimulation using a von Frey hair applied to the plantar surface of the hindpaw. The low-intensity mechanical stimuli increased the discharges during stimulation (during-discharges) without increasing the discharges after cessation of stimulation (after-discharges), and the high-intensity mechanical stimuli increased both during- and after-discharges. Pretreatment with resiniferatoxin, an ultrapotent analogue of capsaicin, halved during- discharges and eliminated after-discharges, suggesting that after-discharges are generated by heat- and mechanosensitive polymodal nociceptors. Ezlopitant, a neurokinin-1 (NK-1) receptor antagonist, but not its inactive enantiomer, selectively reduced the after-discharges. Ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist, preferentially reduced the after-discharges, demonstrating that NK-1 and NMDA receptors mediate the after-discharges. Morphine reduced the after-discharges without affecting during-discharges. By contrast, mephenesin, a centrally acting muscle relaxant, reduced both during- and after-discharges. There results suggest that simultaneous recordings of during- and after-discharges are useful to study pain mechanisms and analgesics as well as to discriminate the analgesic effects from the side effects such as muscle relaxant effects. (c) 2006 Elsevier B.V. All rights reserved.
  • M Tanabe, Y Kino, M Honda, H Ono
    JOURNAL OF NEUROSCIENCE 26(6) 1795-1802 2006年2月  査読有り
    Imidazoline receptors are expressed widely in the CNS. In the present study, whole-cell patch-clamp recordings were made from medium spiny neurons in dorsal striatum slices from the rat brain, and the roles of I-1-imidazoline receptors in the modulation of synaptic transmission were studied. Moxonidine, an I-1-imidazoline receptor agonist, decreased the GABA(A) receptor-mediated IPSCs in a concentration-dependent manner. However, glutamate-mediated EPSCs were hardly affected. The depression of IPSCs by moxonidine was antagonized by either idazoxan or efaroxan, which are both imidazoline receptor antagonists containing an imidazoline moiety. In contrast, yohimbine and SKF86466 (6-chloro-2,3,4,5-tetrahydro-3-methyl-1H-3-benzazepine), which are alpha 2-adrenergic receptor antagonists with no affinity for imidazoline receptors, did not affect the moxonidine-induced inhibition of IPSCs. Moxonidine increased the paired-pulse ratio and reduced the frequency of miniature IPSCs without affecting their amplitude, indicating that this agent inhibits IPSCs via presynaptic mechanisms. Moreover, the sulfhydryl alkylating agent N-ethylmaleimide (NEM) significantly reduced the moxonidine-induced inhibition of IPSCs. Thus, the activation of presynaptic I-1-imidazoline receptors decreases GABA-mediated inhibition of medium spiny neurons in the striatum, in which NEM-sensitive proteins such as G(i/o)-type G-proteins play an essential role. The adenylate cyclase activator forskolin partly opposed IPSC inhibition elicited by subsequently applied moxonidine. Furthermore, the protein kinase C (PKC) activator phorbol 12,13-dibutyrate attenuated and the PKC inhibitor chelerythrine potentiated the moxonidine-induced inhibition of IPSCs. These results suggest that IPSC inhibition via presynaptic I-1-imidazoline receptors involves intracellular adenylate cyclase activity and is influenced by static PKC activity in the striatum.

MISC

 93
  • Masahiro Ohsawa, Shohei Yamamoto, Hideki Ono
    YAKUGAKU ZASSHI-JOURNAL OF THE PHARMACEUTICAL SOCIETY OF JAPAN 134(3) 387-395 2014年3月  
    Central sensitization in the spinal cord is well known to be involved in chronic pain. Recent investigations indicated that the protein expressions involving the synaptic plasticity are changed in several brain areas under a chronic pain condition. These changes in supraspinal neural function might cause the emotional and memory dysfunction. It is also possible that these changes are involved in the chronic pain. Indeed, since the improvement of spinal and peripheral sensitization showed limited relief in the neuropathic pain, the sensitization of supraspinal nociceptive transmission might be involved in the expression of chronic pain. We recently found that intra-thalamic treatment with excitatory neurotransmitter glutamate caused hyperalgesia, which is mediated by the stimulation of glutamate N-methyl-D-aspartate (NMDA) and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Moreover, intracerebroventricular treatment with gabapentin, a calcium channel alpha2delta-1 subunit blocker, attenuated the hyperalgesia in the nerve-injury model of mice. These results suggest that the sensitization of supraspinal nociceptive transmission is involved in neuropathic pain. It is also indicated that neuropathic pain is resulted from the activations of spinal glial cells. Likewise, the supraspinal glial activation was observed in the neuropathic pain. Therefore, the sensitization of supraspinal nociceptive transmission might be important for a chronic pain. In this review, we would like to discuss the possible involvement of the supraspinal sensitization in neuropathic pain and in its application for the curative treatment in chronic pain.
  • Masahiro Ohsawa, Riyo Nakamura, Noboru Inoue, Tomoyasu Murakami, Hiroki Katsu, Shohei Yamamoto, Hideki Ono
    DIABETES 62 A211-A212 2013年7月  
  • Yu Masegi, Hideki Ono, Mitsuo Tanabe
    JOURNAL OF PHARMACOLOGICAL SCIENCES 118 194P-194P 2012年  
  • Seiya Kuraoka, Hideki Ono, Mitsuo Tanabe
    JOURNAL OF PHARMACOLOGICAL SCIENCES 118 193P-193P 2012年  
  • Saki Otake, Shohei Yamamoto, Masahiro Ohsawa, Masahide Noji, Hideki Ono
    JOURNAL OF PHARMACOLOGICAL SCIENCES 118 146P-146P 2012年  

書籍等出版物

 32

講演・口頭発表等

 21

Works(作品等)

 4

共同研究・競争的資金等の研究課題

 20

教育内容・方法の工夫

 2
  • 件名
    認定実務実習指導薬剤師養成ワークショップ in 東海 タスクフォース
    年月日(From)
    2005/12
    年月日(To)
    2008
    概要
    第1回、4回、5回、12回ワークショップにおいて、タスクフォースを務めた。
  • 件名
    文科省戦略的大学連携支援事業「6年制薬学教育を主軸とする薬系・医系・看護系大学による広域総合教育連携(20〜22年度)」 東海4県11大学連携 東海臨床薬学連携センター長
    年月日(From)
    2008/10
    年月日(To)
    2013/03
    概要
    平成20年度から、愛知、岐阜、三重、静岡の11大学の連携において、東海臨床薬学連携センター長として、申請、運営、取りまとめを行った。平成18年度から始まった6年制薬学教育を実施していくために、5つの実施部会を組織し活動を行った。

実務経験を有する者についての特記事項(教育上の能力)

 1
  • 件名
    東京大学医学部附属病院分院薬剤部長
    年月日(From)
    2003/10
    年月日(To)
    2006/04

資格・免許

 1
  • 件名
    薬剤師免許
    年月日
    1976/06/12

実務経験を有する者についての特記事項(職務上の実績)

 1
  • 件名
    東京大学医学部附属病院 分院薬剤部長
    年月日(From)
    1993/10/16
    年月日(To)
    1996/04/25