Network


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

Hotspot


Dive into the research topics where Hideto Morimoto is active.

Publication


Featured researches published by Hideto Morimoto.


Molecular Genetics and Metabolism | 2012

Enzyme replacement therapy (ERT) procedure for mucopolysaccharidosis type II (MPS II) by intraventricular administration (IVA) in murine MPS II.

Takashi Higuchi; Hiromi Shimizu; Shiho Kawagoe; Juri Matsumoto; Yohta Shimada; Hiroshi Kobayashi; Hiroyuki Ida; Toya Ohashi; Hideto Morimoto; Tohru Hirato; Katsuya Nishino; Yoshikatsu Eto

Mucopolysaccharidosis type II (MPS II), or Hunter syndrome, is a lysosomal storage disorder caused by a deficiency of iduronate-2-sulfatase (IDS) and is characterized by the accumulation of glycosaminoglycans (GAGs). MPS II has been treated by hematopoietic stem cell therapy (HSCT)/enzyme replacement therapy (ERT), but its effectiveness in the central nervous system (CNS) is limited because of poor enzyme uptake across the blood-brain barrier (BBB). To increase the efficacy of ERT in the brain, we tested an intraventricular ERT procedure consisting of repeated administrations of IDS (20 μg/mouse/3 weeks) in IDS-knockout, MPS II model mice. The IDS enzyme activity and the accumulation of total GAGs were measured in mouse brains. The IDS activity was significantly increased, and the accumulation of total GAGs was decreased in the MPS II mouse brains treated with multiple administrations of IDS via intraventricular ERT. Additionally, a high level of IDS enzyme activity was appreciated in other MPS II mouse tissues, such as the liver, spleen, testis and others. A Y-maze was used to test learning and memory after repeated intraventricular ERT with IDS. The IDS-treated mouse groups recovered the capacity for short-term memory and activity. Although large and small vacuoles were found at the margin of the cerebellar Purkinje cells in the disease-control mice, these vacuoles disappeared upon treated with IDS. Loss of vacuoles was also observed in other tissues (liver, kidney and testis). These results demonstrate the possible efficacy of an ERT procedure with intraventricular administration of IDS for the treatment of MPS II.


Molecular Therapy | 2018

A blood-brain barrier-penetrating anti-human transferrin receptor antibody fusion protein for neuronopathic mucopolysaccharidosis II

Hiroyuki Sonoda; Hideto Morimoto; Eiji Yoden; Yuri Koshimura; Masafumi Kinoshita; Galina Golovina; Haruna Takagi; Ryuji Yamamoto; Kohtaro Minami; Akira Mizoguchi; Katsuhiko Tachibana; Tohru Hirato; Kenichi Takahashi

Mucopolysaccharidosis II (MPS II) is an X-linked recessive lysosomal storage disease caused by mutations in the iduronate-2-sulfatase (IDS) gene. Since IDS catalyzes the degradation of glycosaminoglycans (GAGs), deficiency in this enzyme leads to accumulation of GAGs in most cells in all tissues and organs, resulting in severe somatic and neurological disorders. Although enzyme replacement therapy with human IDS (hIDS) has been used for the treatment of MPS II, this therapy is not effective for defects in the CNS mainly because the enzyme cannot cross the blood-brain barrier (BBB). Here, we developed a BBB-penetrating fusion protein, JR-141, which consists of an anti-human transferrin receptor (hTfR) antibody and intact hIDS. The TfR-mediated incorporation of JR-141 was confirmed by using human fibroblasts in vitro. When administrated intravenously to hTfR knockin mice or monkeys, JR-141, but not naked hIDS, was detected in the brain. In addition, the intravenous administration of JR-141 reduced the accumulation of GAGs both in the peripheral tissues and in the brain of hTfR knockin mice lacking Ids, an animal model of MPS II. These data provide a proof of concept for the translation of JR-141 to clinical study for the treatment of patients with MPS II with CNS disorders.


Molecular Genetics and Metabolism | 2018

Evaluation of cerebrospinal fluid heparan sulfate as a biomarker of neuropathology in a murine model of mucopolysaccharidosis type II using high-sensitivity LC/MS/MS

Noboru Tanaka; Sachiho Kida; Masafumi Kinoshita; Hideto Morimoto; Tadao Shibasaki; Katsuhiko Tachibana; Ryuji Yamamoto

Mucopolysaccharidosis type II (MPS II or Hunter syndrome) is a lysosomal storage disorder caused by a deficiency of iduronate-2-sulfatase (IDS), an enzyme that catabolizes glycosaminoglycans (GAGs) including heparan sulfate (HS) and dermatan sulfate (DS). GAG accumulation leads to severe neurological and somatic impairments. At present, the most common treatment for MPS II is intravenous enzyme replacement therapy; however, the inability of recombinant IDS to cross the blood-brain barrier (BBB) restricts therapeutic efficacy for neurological manifestations. We recently developed a BBB-penetrating IDS fusion protein, JR-141, and demonstrated its ability to reduce GAG accumulation in the brain of human transferrin receptor knock-in and Ids knock-out mice (TFRC-KI/Ids-KO), an animal model of MPS II, following intravenous administration. Given the impossibility of measuring GAG accumulation in the brains of human patients with MPS II, we hypothesized that GAG content in the cerebrospinal fluid (CSF) might serve as an indicator of brain GAG burden. To test this hypothesis, we optimized a high-sensitivity method for quantifying HS and DS in low-volume samples by combining acidic methanolysis and liquid chromatography-tandem mass spectrometry (LC/MS/MS). We employed this method to quantify HS and DS in samples from TFRC-KI/Ids-KO mice and revealed that HS but not DS accumulated in the central nerve system (CNS). Moreover, concentrations of HS in CSF correlated with those in brain. Finally, intravenous treatment with JR-141 reduced levels of HS in the CSF and brain in TFRC-KI/Ids-KO mice. These results suggest that CSF HS content may be a useful biomarker for evaluating the brain GAG accumulation and the therapeutic efficacy of drugs in patients with MPS II.


Molecular Genetics and Metabolism | 2018

Non-clinical evaluation of JR-051 as a biosimilar to agalsidase beta for the treatment of Fabry disease

Hideto Morimoto; Yae Ito; Eiji Yoden; Masato Horie; Noboru Tanaka; Yoshikazu Komurasaki; Ryuji Yamamoto; Kazutoshi Mihara; Kohtaro Minami; Tohru Hirato

Fabry disease (FD) is an X-linked lysosomal storage disease. It is caused by deficiency of the enzyme α-galactosidase A (α-Gal A), which leads to excessive deposition of neutral glycosphingolipids, especially globotriaosylceramide (GL-3), in cells throughout the body. Progressive accumulation of GL-3 causes life-threatening complications in several tissues and organs, including the vasculature, heart, and kidney. Currently available enzyme replacement therapy for FD employs recombinant α-Gal A in two formulations, namely agalsidase alfa and agalsidase beta. Here, we evaluated JR-051 as a biosimilar to agalsidase beta in a non-clinical study. JR-051 was shown to have identical primary and similar higher-order structures to agalsidase beta. Mannose-6-phosphate content was higher in JR-051 than in agalsidase beta, which probably accounts for a slightly better uptake into fibroblasts in vitro. In spite of these differences in in vitro biological features, pharmacokinetic profiles of the two compounds in mice, rats, and monkeys were similar. The ability to reduce GL-3 accumulation in the kidney, heart, skin, liver, spleen, and plasma of Gla-knockout mice, a model of FD, was not different between JR-051 and agalsidase beta. Furthermore, we identified no safety concerns regarding JR-051 in a 13-week evaluation using cynomolgus monkeys. These findings indicate that JR-051 is similar to agalsidase beta in terms of physicochemical and biological properties.


Archive | 2014

NOVEL ANTI-TRANSFERRIN RECEPTOR ANTIBODY THAT PASSES THROUGH BLOOD-BRAIN BARRIER

啓之 薗田; Hiroyuki Sonoda; 秀人 森本; Hideto Morimoto; 友理 越村; Yuri Koshimura; 正文 木下; Masafumi Kinoshita; 春奈 高木; Haruna Takagi; 佳子 由井; Yoshiko Yoshii


Molecular Genetics and Metabolism | 2017

Correction of central nervous system deficits in the mouse model of Hunter syndrome by recombinant iduronate 2-sulfatase crossing the blood-brain barrier

Hiroyuki Sonoda; Hideto Morimoto; Yuri Koshimura; Masafumi Kinoshita


Molecular Genetics and Metabolism | 2018

Blood-brain barrier-penetrating iduronate-2-sulfatase reduces brain glycosaminoglycans in mouse model of mucopolysaccharidosis type II

Hiroyuki Sonoda; Hideto Morimoto; Eiji Yoden; Yuri Koshimura; Masafumi Kinoshita; Galina Golovina; Haruna Takagi; Ryuji Yamamoto; Kohtaro Minami; Katsuhiko Tachibana; Tohru Hirato; Kenichi Takahashi


Molecular Genetics and Metabolism | 2015

The characterization of mouse model of mucopolysaccharidosis type II

Masafumi Kinoshita; Hideto Morimoto; Eiji Yoden


Molecular Genetics and Metabolism | 2015

Novel quantification methods for globotriaosylceramide and globotriaosylsphingosine as biomarkers of Fabry disease

Noboru Tanaka; Hideto Morimoto; Eiji Yoden; Ryuji Yamamoto


Archive | 2014

Anti-human transferrin receptor antibody that passes through blood-brain barrier

Hiroyuki Sonoda; Hideto Morimoto; Yuri Koshimura; Masafumi Kinoshita; Haruna Takagi; Yoshiko Yoshii

Collaboration


Dive into the Hideto Morimoto's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiromi Shimizu

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hiroshi Kobayashi

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hiroyuki Ida

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Juri Matsumoto

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Shiho Kawagoe

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Takashi Higuchi

Jikei University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge