Network


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

Hotspot


Dive into the research topics where Kiichiro Matsumura is active.

Publication


Featured researches published by Kiichiro Matsumura.


Neurology | 2005

New and reliable MRI diagnosis for progressive supranuclear palsy

H. Oba; A. Yagishita; H. Terada; A. J. Barkovich; K. Kutomi; T. Yamauchi; S. Furui; Teruo Shimizu; M. Uchigata; Kiichiro Matsumura; M. Sonoo; M. Sakai; K. Takada; A. Harasawa; K. Takeshita; H. Kohtake; H. Tanaka; S. Suzuki

OBJECTIVEnTo evaluate the area of the midbrain and pons on mid-sagittal MRI in patients with progressive supranuclear palsy (PSP), Parkinson disease (PD), and multiple-system atrophy of the Parkinson type (MSA-P), compare these appearances and values with those of normal control subjects, and establish diagnostic MRI criteria for the diagnosis of PSP.nnnMETHODSnThe authors prospectively studied MRI of 21 patients with PSP, 23 patients with PD, 25 patients with MSA-P, and 31 age-matched normal control subjects. The areas of the midbrain tegmentum and the pons were measured on mid-sagittal MRI using the display tools of a workstation. The ratio of the area of the midbrain to the area of the pons was also evaluated in all subjects.nnnRESULTSnThe average midbrain area of the patients with PSP (56.0 mm2) was significantly smaller than that of the patients with PD (103.0 mm2) and MSA-P (97.2 mm2) and that of the age-matched control group (117.7 mm2). The values of the area of the midbrain showed no overlap between patients with PSP and patients with PD or normal control subjects. However, patients with MSA-P showed some overlap of the values of individual areas with values from patients with PSP. The ratio of the area of the midbrain to the area of pons in the patients with PSP (0.124) was significantly smaller than that in those with PD (0.208) and MSA-P (0.266) and in normal control subjects (0.237). Use of the ratio allowed differentiation between the PSP group and the MSA-P group.nnnCONCLUSIONnThe area of the midbrain on mid-sagittal MRI can differentiate PSP from PD, MSA-P, and normal aging.


Nature | 2013

LARGE glycans on dystroglycan function as a tunable matrix scaffold to prevent dystrophy

Matthew M. Goddeeris; Biming Wu; David Venzke; Takako Yoshida-Moriguchi; Fumiaki Saito; Kiichiro Matsumura; Steven A. Moore; Kevin P. Campbell

The dense glycan coat that surrounds every cell is essential for cellular development and physiological function, and it is becoming appreciated that its composition is highly dynamic. Post-translational addition of the polysaccharide repeating unit [-3-xylose-α1,3-glucuronic acid-β1-]n by like-acetylglucosaminyltransferase (LARGE) is required for the glycoprotein dystroglycan to function as a receptor for proteins in the extracellular matrix. Reductions in the amount of [-3-xylose-α1,3-glucuronic acid-β1-]n (hereafter referred to as LARGE-glycan) on dystroglycan result in heterogeneous forms of muscular dystrophy. However, neither patient nor mouse studies has revealed a clear correlation between glycosylation status and phenotype. This disparity can be attributed to our lack of knowledge of the cellular function of the LARGE-glycan repeat. Here we show that coordinated upregulation of Large and dystroglycan in differentiating mouse muscle facilitates rapid extension of LARGE-glycan repeat chains. Using synthesized LARGE-glycan repeats we show a direct correlation between LARGE-glycan extension and its binding capacity for extracellular matrix ligands. Blocking Large upregulation during muscle regeneration results in the synthesis of dystroglycan with minimal LARGE-glycan repeats in association with a less compact basement membrane, immature neuromuscular junctions and dysfunctional muscle predisposed to dystrophy. This was consistent with the finding that patients with increased clinical severity of disease have fewer LARGE-glycan repeats. Our results reveal that the LARGE-glycan of dystroglycan serves as a tunable extracellular matrix protein scaffold, the extension of which is required for normal skeletal muscle function.


Journal of Neurology, Neurosurgery, and Psychiatry | 2005

Age associated axonal features in HNPP with 17p11.2 deletion in Japan

Haruki Koike; Masaaki Hirayama; Masahiko Yamamoto; H Ito; Naoki Hattori; Fujio Umehara; Kimiyoshi Arimura; Shinji Ikeda; Yukio Ando; Masamitsu Nakazato; Ryuji Kaji; Kiyoshi Hayasaka; Masanori Nakagawa; Saburo Sakoda; Kiichiro Matsumura; Osamu Onodera; Masayuki Baba; Hitoshi Yasuda; Toyokazu Saito; Jun-ichi Kira; Kenji Nakashima; Nobuyuki Oka; Gen Sobue

Objective: To clarify age related changes in the clinicopathological features of hereditary neuropathy with liability to pressure palsy (HNPP) in Japanese patients with deletion of 17p11.2, particularly concerning axonal abnormalities. Methods: Forty eight proband patients from 48 HNPP families were assessed as to clinical, electrophysiological, and histopathological features, including age associated changes beyond those in controls. Results: Motor conduction studies showed age associated deterioration of compound muscle action potentials in nerves vulnerable to repetitive compression (median, ulnar, and peroneal nerves), but not in others such as the tibial nerve. Sensory conduction studies revealed more profound reduction of action potentials than motor studies with little age related change. Large myelinated fibre loss was seen in the sural nerve irrespective of age at examination. Conclusions: Irreversible axonal damage may occur at entrapment sites in motor nerves in HNPP patients, progressing with aging. Sensory nerves may show more profound axonal abnormality, but without age association. The electrophysiological features of HNPP are presumed to be a mixture of abnormalities occurring from early in life and acquired features caused by repetitive insults at entrapment sites. Unlike Charcot-Marie-Tooth disease type 1A, age associated axonal damage may not occur unless the nerves are subjected to compression.


Neuromuscular Disorders | 2005

Proteolysis of β-dystroglycan in muscular diseases

Kiichiro Matsumura; Di Zhong; Fumiaki Saito; Ken Arai; Katsuhito Adachi; Hisaomi Kawai; Itsuro Higuchi; Ichizo Nishino; Teruo Shimizu

Alpha-dystroglycan is a cell surface peripheral membrane protein which binds to the extracellular matrix (ECM), while beta-dystroglycan is a type I integral membrane protein which anchors alpha-dystroglycan to the cell membrane via the N-terminal extracellular domain. The complex composed of alpha-and beta-dystroglycan is called the dystroglycan complex. We reported previously a matrix metalloproteinase (MMP) activity that disrupts the dystroglycan complex by cleaving the extracellular domain of beta-dystroglycan. This MMP creates a characteristic 30 kDa fragment of beta-dystroglycan that is detected by the monoclonal antibody 43DAG/8D5 directed against the C-terminus of beta-dystroglycan. We also reported that the 30 kDa fragment of beta-dystroglycan was increased in the skeletal and cardiac muscles of cardiomyopathic hamsters, the model animals of sarcoglycanopathy, and that this resulted in the disruption of the link between the ECM and cell membrane via the dystroglycan complex. In this study, we investigated the proteolysis of beta-dystroglycan in the biopsied skeletal muscles of various human muscular diseases, including sarcoglycanopathy, Duchenne muscular dystrophy (DMD), Becker muscular dystrophy, Fukuyama congenital muscular dystrophy, Miyoshi myopathy, LGMD2A, facioscapulohumeral muscular dystrophy, myotonic dystrophy and dermatomyositis/polymyositis. We show that the 30 kDa fragment of beta-dystroglycan is increased significantly in sarcoglycanopathy and DMD, but not in the other diseases. We propose that the proteolysis of beta-dystroglycan may contribute to skeletal muscle degeneration by disrupting the link between the ECM and cell membrane in sarcoglycanopathy and DMD.


FEBS Letters | 2005

Aberrant glycosylation of α‐dystroglycan causes defective binding of laminin in the muscle of chicken muscular dystrophy

Fumiaki Saito; Martina Blank; Jörn E. Schröder; Hiroshi Manya; Teruo Shimizu; Kevin P. Campbell; Tamao Endo; Makoto Mizutani; Stephan Kröger; Kiichiro Matsumura

Dystroglycan is a central component of dystrophin–glycoprotein complex that links extracellular matrix and cytoskeleton in skeletal muscle. Although dystrophic chicken is well established as an animal model of human muscular dystrophy, the pathomechanism leading to muscular degeneration remains unknown. We show here that glycosylation and laminin‐binding activity of α‐dystroglycan (α‐DG) are defective in dystrophic chicken. Extensive glycan structural analysis reveals that Galβ1‐3GalNAc and GalNAc residues are increased while Siaα2‐3Gal structure is reduced in α‐DG of dystrophic chicken. These results implicate aberrant glycosylation of α‐DG in the pathogenesis of muscular degeneration in this model animal of muscular dystrophy.


Journal of Neurochemistry | 2007

Defective peripheral nerve myelination and neuromuscular junction formation in fukutin-deficient chimeric mice

Fumiaki Saito; Toshihiro Masaki; Yuko Saito; Ayami Nakamura; Satoshi Takeda; Teruo Shimizu; Tatsushi Toda; Kiichiro Matsumura

Dystroglycan is a central component of the dystrophin–glycoprotein complex that links the extracellular matrix with cytoskeleton. Recently, mutations of the genes encoding putative glycosyltransferases were identified in several forms of congenital muscular dystrophies accompanied by brain anomalies and eye abnormalities, and aberrant glycosylation of α‐dystroglycan has been implicated in their pathogeneses. These diseases are now collectively called α‐dystroglycanopathy. In this study, we demonstrate that peripheral nerve myelination is defective in the fukutin‐deficient chimeric mice, a mouse model of Fukuyama‐type congenital muscular dystrophy, which is the most common α‐dystroglycanopathy in Japan. In the peripheral nerve of these mice, the density of myelinated nerve fibers was significantly decreased and clusters of abnormally large non‐myelinated axons were ensheathed by a single Schwann cell, indicating a defect of the radial sorting mechanism. The sugar chain moiety and laminin‐binding activity of α‐dystroglycan were severely reduced, while the expression of β1‐integrin was not altered in the peripheral nerve of the chimeric mice. We also show that the clustering of acetylcholine receptor is defective and neuromuscular junctions are fragmented in appearance in these mice. Expression of agrin and laminin as well as the binding activity of α‐dystroglycan to these ligands was severely reduced at the neuromuscular junction. These results demonstrate that fukutin plays crucial roles in the myelination of peripheral nerve and formation of neuromuscular junction. They also suggest that defective glycosylation of α‐dystroglycan may play a role in the impairment of these processes in the deficiency of fukutin.


BioMed Research International | 2010

Biological Role of Dystroglycan in Schwann Cell Function and Its Implications in Peripheral Nervous System Diseases

Toshihiro Masaki; Kiichiro Matsumura

Dystroglycan is a central component of the dystrophin-glycoprotein complex (DGC) that links extracellular matrix with cytoskeleton, expressed in a variety of fetal and adult tissues. Dystroglycan plays diverse roles in development and homeostasis including basement membrane formation, epithelial morphogenesis, membrane stability, cell polarization, and cell migration. In this paper, we will focus on biological role of dystroglycan in Schwann cell function, especially myelination. First, we review the molecular architecture of DGC in Schwann cell abaxonal membrane. Then, we will review the loss-of-function studies using targeted mutagenesis, which have revealed biological functions of each component of DGC in Schwann cells. Based on these findings, roles of dystroglycan in Schwann cell function, in myelination in particular, and its implications in diseases will be discussed in detail. Finally, in view of the fact that understanding the role of dystroglycan in Schwann cells is just beginning, future perspectives will be discussed.


FEBS Letters | 2008

Processing and secretion of the N‐terminal domain of α‐dystroglycan in cell culture media

Fumiaki Saito; Yuko Saito-Arai; Ayami Nakamura; Teruo Shimizu; Kiichiro Matsumura

α‐Dystroglycan (α‐DG) plays a crucial role in maintaining the stability of muscle cell membrane. Although it has been shown that the N‐terminal domain of α‐DG (α‐DG‐N) is cleaved by a proprotein convertase, its physiological significance remains unclear. We show here that native α‐DG‐N is secreted by a wide variety of cultured cells into the culture media. The secreted α‐DG‐N was both N‐ and O‐glycosylated. Finally, a small amount of α‐DG‐N was detectable in the normal human serum. These observations indicate that the cleavage of α‐DG‐N is a widespread event and suggest that the secreted α‐DG‐N might be transported via systemic circulation in vivo.


Kidney International | 2011

Defective glycosylation of α-dystroglycan contributes to podocyte flattening

Kenichiro Kojima; Hitonari Nosaka; Yuki Kishimoto; Yuri Nishiyama; Seiichi Fukuda; Masaru Shimada; Kenzo Kodaka; Fumiaki Saito; Kiichiro Matsumura; Teruo Shimizu; Tatsushi Toda; Satoshi Takeda; Hiroshi Kawachi; Shunya Uchida

In addition to skeletal muscle and the nervous system, α-dystroglycan is found in the podocyte basal membrane, stabilizing these cells on the glomerular basement membrane. Fukutin, named after the gene responsible for Fukuyama-type congenital muscular dystrophy, is a putative glycosyltransferase required for the post-translational modification of α-dystroglycan. Chimeric mice targeted for both alleles of fukutin develop severe muscular dystrophy; however, these mice do not have proteinuria. Despite the lack of a functional renal defect, we evaluated glomerular structure and found minor abnormalities in the chimeric mice by light microscopy. Electron microscopy revealed flattening of podocyte foot processes, the number of which was significantly lower in the chimeric compared to wild-type mice. A monoclonal antibody against the laminin-binding carbohydrate residues of α-dystroglycan did not detect α-dystroglycan glycosylation in the glomeruli by immunoblotting or immunohistochemistry. In contrast, expression of the core α-dystroglycan protein was preserved. There was no statistical difference in dystroglycan mRNA expression or in the amount of nephrin and α3-integrin protein in the chimeric compared to the wild-type mice as judged by immunohistochemistry and real-time RT-PCR. Thus, our results indicate that appropriate glycosylation of α-dystroglycan has an important role in the maintenance of podocyte architecture.


Human Molecular Genetics | 2014

Overexpression of LARGE suppresses muscle regeneration via down-regulation of insulin-like growth factor 1 and aggravates muscular dystrophy in mice

Fumiaki Saito; Motoi Kanagawa; Miki Ikeda; Hiroki Hagiwara; Toshihiro Masaki; Hidehiko Ohkuma; Yuki Katanosaka; Teruo Shimizu; Masahiro Sonoo; Tatsushi Toda; Kiichiro Matsumura

Several types of muscular dystrophy are caused by defective linkage between α-dystroglycan (α-DG) and laminin. Among these, dystroglycanopathy, including Fukuyama-type congenital muscular dystrophy (FCMD), results from abnormal glycosylation of α-DG. Recent studies have shown that like-acetylglucosaminyltransferase (LARGE) strongly enhances the laminin-binding activity of α-DG. Therefore, restoration of the α-DG-laminin linkage by LARGE is considered one of the most promising possible therapies for muscular dystrophy. In this study, we generated transgenic mice that overexpress LARGE (LARGE Tg) and crossed them with dy(2J) mice and fukutin conditional knockout mice, a model for laminin α2-deficient congenital muscular dystrophy (MDC1A) and FCMD, respectively. Remarkably, in both the strains, the transgenic overexpression of LARGE resulted in an aggravation of muscular dystrophy. Using morphometric analyses, we found that the deterioration of muscle pathology was caused by suppression of muscle regeneration. Overexpression of LARGE in C2C12 cells further demonstrated defects in myotube formation. Interestingly, a decreased expression of insulin-like growth factor 1 (IGF-1) was identified in both LARGE Tg mice and LARGE-overexpressing C2C12 myotubes. Supplementing the C2C12 cells with IGF-1 restored the defective myotube formation. Taken together, our findings indicate that the overexpression of LARGE aggravates muscular dystrophy by suppressing the muscle regeneration and this adverse effect is mediated via reduced expression of IGF-1.

Collaboration


Dive into the Kiichiro Matsumura's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin P. Campbell

Roy J. and Lucille A. Carver College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge