Network


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

Hotspot


Dive into the research topics where Hiide Yoshino is active.

Publication


Featured researches published by Hiide Yoshino.


Neurology | 1990

Acute axonal polyneuropathy associated with anti‐GM1 antibodies following Campylobacter enteritis

Nobuhiro Yuki; Hiide Yoshino; Shuzo Sato; Tadashi Miyatake

We report 2 patients with Guillain-Barré syndrome (GBS) following Campylobacter jejuni enteritis. Electrophysiologic studies indicated that the predominant process was axonal degeneration of motor nerves, and clinical recovery was poor. Serum testing by thin-layer chromatography and enzyme-linked immunosorbent assay revealed that the sera from both patients contained high titers of IgG antibody against GM1 ganglioside. These cases may represent a subgroup of GBS as acute axonal polyneuropathy following C jejuni enteritis associated with anti-GM1 antibodies.


Amyotrophic Lateral Sclerosis | 2006

Investigation of the therapeutic effects of edaravone, a free radical scavenger, on amyotrophic lateral sclerosis (Phase II study).

Hiide Yoshino; Akio Kimura

Amyotrophic lateral sclerosis (ALS) is a rare disease involving selective and progressive degeneration and disappearance of motor neurons. Oxidative stress is believed to contribute to its pathogenesis. We have investigated the efficacy and safety of edaravone, a free radical scavenger previously approved for treatment of acute cerebral infarction, in ALS patients. Within an open trial design, 20 subjects with ALS received either 30 mg (5 subjects) or 60 mg (15 subjects) of edaravone via intravenous drip once per day. Two weeks of administration was followed by a two‐week observation period. This four‐week cycle was repeated six times. The primary endpoint was the change in the revised ALS functional rating scale (ALSFRS‐R) score, while the secondary endpoint was 3‐nitrotyrosine (3NT) level in cerebrospinal fluid (CSF). Efficacy was evaluated in the 60 mg group. During the six‐month treatment period, the decline in the ALSFRS‐R score (2.3±3.6 points) was significantly less than that in the six months prior to edaravone administration (4.7±2.1 points); the difference between the two was 2.4±3.5 points (Wilcoxon signed rank test, p = 0.039). In almost all patients, CSF 3NT, a marker for oxidative stress, was markedly reduced to almost undetectable levels at the end of the six‐month treatment period. Data from the present study suggest that edaravone is safe and may delay the progression of functional motor disturbances by reducing oxidative stress in ALS patients.


Lancet Neurology | 2017

Safety and efficacy of edaravone in well defined patients with amyotrophic lateral sclerosis: a randomised, double-blind, placebo-controlled trial

Koji Abe; Masashi Aoki; Shoji Tsuji; Yasuto Itoyama; Gen Sobue; Masanori Togo; Chikuma Hamada; Masahiko Tanaka; Makoto Akimoto; Kazue Nakamura; Fumihiro Takahashi; Kazuoki Kondo; Hiide Yoshino; Hidenao Sasaki; Ichiro Yabe; Shizuki Doi; Hitoshi Warita; Takashi Imai; Hiroaki Ito; Mitsumasa Fukuchi; Etsuko Osumi; Manabu Wada; Imaharu Nakano; Mitsuya Morita; Katsuhisa Ogata; Yuichi Maruki; Kimiko Ito; Osamu Kano; Mineo Yamazaki; Yuji Takahashi

BACKGROUND In a previous phase 3 study in patients with amyotrophic lateral sclerosis (ALS), edaravone did not show a significant difference in the Revised ALS Functional Rating Scale (ALSFRS-R) score compared with placebo. Post-hoc analysis of these data revealed that patients in an early stage with definite or probable diagnosis of ALS, defined by the revised El Escorial criteria, who met a select set of inclusion criteria showed a greater magnitude of effect than did the full study population. We aimed to substantiate this post-hoc result and assess safety and efficacy of edaravone in a phase 3 trial that focused on patients with early stage ALS who met the post-hoc analysis inclusion criteria. METHODS In this phase 3, randomised, double-blind, parallel-group study, patients aged 20-75 years with ALS of grade 1 or 2 in the Japan ALS Severity Classification, scores of at least 2 points on all 12 items of ALSFRS-R, forced vital capacity of 80% or more, definite or probable ALS according to the revised El Escorial criteria, and disease duration of 2 years or less were recruited from 31 hospitals in Japan. Eligible patients also had a decrease of 1-4 points in the ALSFRS-R score during a 12-week observation period before randomisation. Patients meeting all criteria were then randomly assigned 1:1 to receive 60 mg intravenous edaravone or intravenous saline placebo for 6 cycles (4 weeks per cycle with 2 weeks on, 2 weeks off) for a total treatment duration of 24 weeks. In cycle 1, the study drug or placebo was administered once per day for 14 days within a 14 day period, followed by the drug-free period. In cycle 2 and thereafter, the study drug or placebo was administered for 10 days within a 14 day period, followed by a 2 week drug-free period. Participants and investigators, including those assessing outcomes, were masked to treatment allocation. The primary efficacy outcome was the change in ALSFRS-R score from the baseline to 24 weeks (or at discontinuation if this was after the third cycle) after randomisation. The primary outcome was assessed in all patients who had received at least one treatment infusion, had at least one assessment post-baseline, and reached the end of cycle 3. For patients with missing values at the end of cycle 6, data were imputed by the last observation carried forward (LOCF) method, provided the patients had completed at least cycle 3. Safety was assessed in all patients who had received at least one treatment infusion and had at least one assessment post-baseline. This trial is registered with ClinicalTrials.gov, NCT01492686. FINDINGS Between Nov 28, 2011, and Sept 3, 2014, we screened 213 patients, and enrolled 192 as potential participants. Of these, 137 patients completed the observation period: 69 were randomly assigned to receive edaravone, and 68 were randomly assigned to receive placebo. 68 patients taking edaravone and 66 taking placebo were included in the primary efficacy analysis. For the primary outcome, the change in ALSFRS-R score was -5·01 (SE 0·64) in the edavarone group and -7·50 (0·66) in the placebo group. The least-squares mean difference between groups was 2·49 (SE 0·76, 95% CI 0·99-3·98; p=0·0013) in favour of edaravone. Treatment-emergent adverse events were reported in 58 (84%) patients receiving edaravone and 57 (84%) patients receiving placebo. 11 (16%) patients taking edaravone and 16 (24%) taking placebo had serious adverse events, and one (1%) patient receiving edaravone and four (6%) patients receiving placebo had adverse events (one dysphagia in edaravone group and one dyspnoea, two respiratory disorder, and one rash in the placebo group) that led to withdrawal. INTERPRETATION Edaravone showed efficacy in a small subset of people with ALS who met criteria identified in post-hoc analysis of a previous phase 3 study, showing a significantly smaller decline of ALSFRS-R score compared with placebo. There is no indication that edaravone might be effective in a wider population of patients with ALS who do not meet the criteria. FUNDING Mitsubishi Tanabe Pharma Corporation.Summary Background In a previous phase 3 study in patients with amyotrophic lateral sclerosis (ALS), edaravone did not show a significant difference in the Revised ALS Functional Rating Scale (ALSFRS-R) score compared with placebo. Post-hoc analysis of these data revealed that patients in an early stage with definite or probable diagnosis of ALS, defined by the revised El Escorial criteria, who met a select set of inclusion criteria showed a greater magnitude of effect than did the full study population. We aimed to substantiate this post-hoc result and assess safety and efficacy of edaravone in a phase 3 trial that focused on patients with early stage ALS who met the post-hoc analysis inclusion criteria. Methods In this phase 3, randomised, double-blind, parallel-group study, patients aged 20–75 years with ALS of grade 1 or 2 in the Japan ALS Severity Classification, scores of at least 2 points on all 12 items of ALSFRS-R, forced vital capacity of 80% or more, definite or probable ALS according to the revised El Escorial criteria, and disease duration of 2 years or less were recruited from 31 hospitals in Japan. Eligible patients also had a decrease of 1–4 points in the ALSFRS-R score during a 12-week observation period before randomisation. Patients meeting all criteria were then randomly assigned 1:1 to receive 60 mg intravenous edaravone or intravenous saline placebo for 6 cycles (4 weeks per cycle with 2 weeks on, 2 weeks off) for a total treatment duration of 24 weeks. In cycle 1, the study drug or placebo was administered once per day for 14 days within a 14 day period, followed by the drug-free period. In cycle 2 and thereafter, the study drug or placebo was administered for 10 days within a 14 day period, followed by a 2 week drug-free period. Participants and investigators, including those assessing outcomes, were masked to treatment allocation. The primary efficacy outcome was the change in ALSFRS-R score from the baseline to 24 weeks (or at discontinuation if this was after the third cycle) after randomisation. The primary outcome was assessed in all patients who had received at least one treatment infusion, had at least one assessment post-baseline, and reached the end of cycle 3. For patients with missing values at the end of cycle 6, data were imputed by the last observation carried forward (LOCF) method, provided the patients had completed at least cycle 3. Safety was assessed in all patients who had received at least one treatment infusion and had at least one assessment post-baseline. This trial is registered with ClinicalTrials.gov, NCT01492686. Findings Between Nov 28, 2011, and Sept 3, 2014, we screened 213 patients, and enrolled 192 as potential participants. Of these, 137 patients completed the observation period: 69 were randomly assigned to receive edaravone, and 68 were randomly assigned to receive placebo. 68 patients taking edaravone and 66 taking placebo were included in the primary efficacy analysis. For the primary outcome, the change in ALSFRS-R score was −5·01 (SE 0·64) in the edavarone group and −7·50 (0·66) in the placebo group. The least-squares mean difference between groups was 2·49 (SE 0·76, 95% CI 0·99–3·98; p=0·0013) in favour of edaravone. Treatment-emergent adverse events were reported in 58 (84%) patients receiving edaravone and 57 (84%) patients receiving placebo. 11 (16%) patients taking edaravone and 16 (24%) taking placebo had serious adverse events, and one (1%) patient receiving edaravone and four (6%) patients receiving placebo had adverse events (one dysphagia in edaravone group and one dyspnoea, two respiratory disorder, and one rash in the placebo group) that led to withdrawal. Interpretation Edaravone showed efficacy in a small subset of people with ALS who met criteria identified in post-hoc analysis of a previous phase 3 study, showing a significantly smaller decline of ALSFRS-R score compared with placebo. There is no indication that edaravone might be effective in a wider population of patients with ALS who do not meet the criteria. Funding Mitsubishi Tanabe Pharma Corporation.


Annals of Neurology | 1999

Anti-GalNAc-GD1a antibody–associated Guillain-Barré syndrome with a predominantly distal weakness without cranial nerve impairment and sensory disturbance

Qi Hao; Takahiko Saida; Hiide Yoshino; Kuroki S; Masafumi Nukina; Kyoko Saida

The serum antibodies to N‐acetylgalactosaminyl GD1a (GalNAc‐GD1a) and other gangliosides as well as to Campylobacter jejuni were determined in 147 patients with Guillain‐Barré syndrome (GBS). We found a distinctive clinical pattern in patients with anti‐GalNAc‐GD1a antibodies compared with those without the antibodies, that is, lack of cranial nerve involvement (87% versus 38%), distal‐dominant weakness (80% versus 25%), and no sensory disturbance (73% versus 22%). The frequency of distal‐dominant weakness was significantly higher in patients with both C jejuni infection and anti‐GalNAc‐GD1a positivity (100%) than in C jejuni–negative/anti‐GalNAc‐GD1a–positive (25%), C jejuni–positive/anti‐GalNAc‐GD1a–negative (32%) and C jejuni–negative/anti‐GalNAc‐GD1a–negative patients (20%). Lack of cranial nerve involvement and sensory disturbance were found in most C jejuni–positive/anti‐GalNAc‐GD1a–positive and C jejuni–negative/anti‐GalNAc‐GD1a–positive patients, but not in C jejuni–positive/anti‐GalNAc‐GD1a–negative and C jejuni–negative/anti‐GalNAc‐GD1a–negative patients. Although the anti‐GM1–positive/anti‐GalNAc‐GD1a–negative patients mostly (75%) lacked cranial nerve involvement, distal‐dominant weakness (38%) and lack of sensory disturbance (13%) were infrequent. These results may indicate that (1) the combination of C jejuni infection and anti‐GalNAc‐GD1a antibodies, but not anti‐GalNAc‐GD1a, anti‐GM1, or C jejuni infection alone, is associated with a predominantly distal weakness, (2) the presence of anti‐GalNAc‐GD1a, rather than C jejuni infection or anti‐GM1 antibody, is associated with a lack of sensory disturbance, (3) both anti‐GalNAc‐GD1a and anti‐GM1 antibodies are independently associated with a lack of cranial nerve impairment. Ann Neurol 1999;45:758–768


Amyotrophic Lateral Sclerosis | 2014

Confirmatory double-blind, parallel-group, placebo-controlled study of efficacy and safety of edaravone (MCI-186) in amyotrophic lateral sclerosis patients

Koji Abe; Yasuto Itoyama; Gen Sobue; Shoji Tsuji; Masashi Aoki; Manabu Doyu; Chikuma Hamada; Kazuoki Kondo; Takatomo Yoneoka; Makoto Akimoto; Hiide Yoshino

Abstract Our objective was to confirm the efficacy and safety of edaravone in amyotrophic lateral sclerosis (ALS) patients. We conducted a 36-week confirmatory study, consisting of 12-week pre-observation period followed by 24-week treatment period. Patients received placebo or edaravone i.v. infusion over 60 min for the first 14 days in cycle 1, and for 10 of the first 14 days during cycles 2 to 6. The efficacy primary endpoint was changed in the revised ALS functional rating scale (ALSFRS-R) scores during the 24-week treatment. Patients were treated with placebo (n = 104) and edaravone (n = 102). Changes in ALSFRS-R during the 24-week treatment were −6.35 ± 0.84 in the placebo group (n = 99) and −5.70 ± 0.85 in the edaravone group (n = 100), with a difference of 0.65 ± 0.78 (p = 0.411). Adverse events amounted to 88.5% (92/104) in the placebo group and 89.2% (91/102) in the edaravone group. In conclusion, the reduction of ALSFRS-R was smaller in the edaravone group than in the placebo group, but efficacy of edaravone for treatment of ALS was not demonstrated. Levels and frequencies of reported adverse events were similar in the two groups. Trial registration: ClinicalTrials.gov identifier: NCT00330681.


Journal of Neurology, Neurosurgery, and Psychiatry | 2002

Anti-GT1a IgG in Guillain-Barré syndrome

Michiaki Koga; Hiide Yoshino; Mitsunori Morimatsu; Nobuhiro Yuki

Objective: To investigate the presence of serum anti-GT1a IgG in Guillain-Barré syndrome (GBS) and its relation to clinical manifestations. Background: Several patients with GBS and bulbar palsy have been reported to have serum anti-GT1a IgG. Most, however, also have anti-GQ1b IgG. A previous study failed to detect GT1a in human cranial nerves, but GQ1b was abundant in human ocular motor nerves. Whether anti-GT1a IgG itself determines the clinical manifestations is not yet clear. Methods: The association of clinical manifestations with the presence of anti-GT1a IgG and with its cross reactivity was investigated. An immunochemical study was performed to determine whether GT1a is present in human cranial nerves. Results: Anti-GT1a and anti-GQ1b IgG were positive in 10% and 9% respectively of 220 consecutive patients with GBS. Patients with anti-GT1a IgG often had cranial nerve palsy (ophthalmoparesis, 57%; facial palsy, 57%; bulbar palsy, 70%), and 39% needed artificial ventilation. These features were also seen in patients with anti-GQ1b IgG. There was no significant difference between the two groups with respect to the frequency of clinical findings. An enzyme-linked immunosorbent assay showed that anti-GT1a IgG cross reacted with GQ1b in 75% of the patients, GD1a in 30%, GM1 in 20%, and GD1b in 20%. All five patients who carried anti-GT1a IgG that did not cross react with GQ1b had bulbar palsy, neck weakness, absence of sensory disturbance, and positive Campylobacter jejuni serology. Thin-layer chromatography with immunostaining showed that GT1a is present in human oculomotor and lower cranial nerves. Conclusions: These findings provide further evidence that anti-GT1a IgG itself can determine clinical manifestations. The distinctive clinical features of patients with anti-GT1a IgG without anti-GQ1b activity distinguish a specific subgroup within GBS.


Journal of the Neurological Sciences | 2004

Neurophysiological and immunohistochemical studies on Guillain-Barré syndrome with IgG anti-GalNAc-GD1a antibodies—effects on neuromuscular transmission

Kyoji Taguchi; Jin Ren; Iku Utsunomiya; Hiroyuki Aoyagi; Nobuya Fujita; Toshio Ariga; Tadashi Miyatake; Hiide Yoshino

We investigated the epitopes and functional role of IgG anti-GalNAc-GD1a antibodies appearing in serum from a patient with Guillain-Barre syndrome (GBS) and IgG anti-GalNAc-GD1a antibody that was produced by immunization of a rabbit with GalNAc-GD1a. Both sera blocked neuromuscular transmission in muscle-spinal cord co-culture cells. The acetylcholine-induced potential did not reduce by adding sera, suggesting that the blockade is presynaptic. The effect was complement-independent and purified IgG from serum of the patient or the rabbit had the same effects. The epitope with both anti-GalNAc-GD1a antibodies was observed in the soma of large neurons in the anterior horns of the adult rat spinal cord and their motor axons of rat ventral roots. Both anti-GalNAc GD1a antibodies reacted strongly with the motor nerve terminals in rats. The anti-GalNAc-GD1a antibodies may block neuromuscular transmission by attacking on presynaptic motor axon, probably affecting the ion channels in the presynaptic motor axon.


Journal of the Neurological Sciences | 1995

Ganglioside characterization of a cell line displaying motor neuron-like phenotype : GM2 as a possible major ganglioside in motor neurons

Akiko Matsumoto; Hiide Yoshino; Nobuhiro Yuki; Yukichi Hara; Neil R. Cashman; Shizuo Handa; Tadashi Miyatake

We have examined ganglioside compositions and the presence of sulfated glucuronyl glycolipids of immortalized motor neuron-like cell lines, neuroblastoma-spinal cord (NSC) hybrid cell lines established by fusing mouse neuroblastoma N18TG2 with motor neuron-enriched embryonic spinal cord cells. Among NSC cell lines, only NSC-34 aggregates acetylcholine receptors on co-cultured myotube and expresses a receptor for S-laminin, a neuromuscular junction specific basal lamina protein. GM2, which is only a minor ganglioside component of CNS, was the major component in NSC-34 occupying almost 75% of total gangliosides, whereas GD1a and GM3 were major species in the parental N18TG2, which had only 8.5% GM2. These results indicated that NSC lines have unique ganglioside pattern that is distinctive from other nervous tissues, and this pattern, especially that of NSC-34 cells, might reflect the characteristics of mouse spinal motor neuron gangliosides. Sulfated glucuronyl paragloboside was demonstrated to be present in N18TG2, however, it could not be detected in either of NSC cell lines. Even though the pathogenesis of amyotrophic lateral sclerosis remains unknown, autoimmunological participation has been suggested. Because high-titered antibody against GM2 has been observed in a patient with amyotrophic lateral sclerosis-like disease, GM2 which is possibly expressed on the surface of motor neurons might serve as a potential target antigen in this disorder.


Journal of Neuroimmunology | 2000

´ IgG antiganglioside antibodies in Guillain-Barre syndrome with bulbar palsy

Hiide Yoshino; Hajime Harukawa; Atsuko Asano

Some patients with Guillain-Barré syndrome (GBS) develop bulbar palsy, which may lead to serious complications during the acute phase of the illness. A serological marker that could predict the occurrence of bulbar palsy would be valuable for the treatment of acute GBS. We examined the serum levels of various IgG antiganglioside antibodies in the sera of 16 patients with GBS with bulbar palsy [GBS-BP(+)] and 72 patients with GBS without bulbar palsy [GBS-BP(-)]. Anti-GT1a antibodies were detected in a higher percentage of the GBS-BP(+) patients (10/16, 63%) than the GBS-BP(-) patients (2/72, 3%). In addition to GT1a, a new disialosylganglioside antigen was recognized by the sera of four GBS-BP(+) patients. Anti-GM1b antibodies were also frequently detected in the sera of the GBS-BP(+) cases. However, anti-GM1 and anti-GalNAc-GD1a antibodies, which are highly associated with acute axonal motor neuropathy (AMAN), were not detected in any of the GBS-BP(+) cases, while anti-GM1 antibodies were detected in 29% (21/72) and anti-GalNAc-GD1a antibodies were detected in 8% (6/72) of the GBS-BP(-) cases. These findings suggest that the presence of particular antiganglioside antibodies might be related with certain clinical manifestations of GBS. In patients who are diagnosed with GBS, the presence or absence of anti-GT1a and anti-GM1b antibodies should be tested at the early stage of GBS so that appropriate therapies that prevent the development of bulbar palsy and improve the outcome of GBS, may be initiated.


Journal of the Neurological Sciences | 1990

Abnormal glycosphingolipid metabolism in the nervous system of galactosialidosis

Hiide Yoshino; Kotaro Miyashita; Nobuyuki Miyatani; Toshio Ariga; Yasuhiro Hashimoto; Shoji Tsuji; Kiyomitsu Oyanagi; E. Ohama; F. Ikuta; Akemi Suzuki; Tadashi Miyatake

In an autopsy case of galactosialidosis, GM3, GM2, GM1, and GD1a were accumulated in sympathetic and spinal ganglia and grey matter of the spinal cord. Especially, the accumulations of GM3 and GM2 amounted to 41- and 86-fold increases in sympathetic ganglia, respectively, as compared to normal controls. In addition LacCer, GA2 and GA1 were accumulated in sympathetic and spinal ganglia. The accumulations of GM3 and GD1a are considered to be the result of defective lysosomal sialidase activity and the accumulation of GM1, LacCer and GA1 is also considered to be due to decreased beta-galactosidase activity in this disorder. To better understand the possible mechanism of GM2 accumulation, we determined the activity of GM2 synthesizing enzyme (GM3:UDP-GalNAc transferase), as well as hexosaminidase activity, in sympathetic ganglia, but they did not change. Abnormal ganglioside and neutral glycosphingolipid metabolism, as well as sialyloligosaccharide and sialylglycoprotein metabolism, may be involved in the pathogenesis of this disorder.

Collaboration


Dive into the Hiide Yoshino's collaboration.

Top Co-Authors

Avatar

Tadashi Miyatake

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Nobuhiro Yuki

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Atsuko Asano

University of Tokushima

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Takao Mitsui

University of Tokushima

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Iku Utsunomiya

Showa Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Kyoji Taguchi

Showa Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge