Network


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

Hotspot


Dive into the research topics where Kazunori Sango is active.

Publication


Featured researches published by Kazunori Sango.


Journal of Biological Chemistry | 2001

Mice Expressing Only Monosialoganglioside GM3 Exhibit Lethal Audiogenic Seizures

Hiromichi Kawai; Maria L. Allende; Ryuichi Wada; Mari Kono; Kazunori Sango; Chu-Xia Deng; Tsuyoshi Miyakawa; Jacqueline N. Crawley; Norbert Werth; Uwe Bierfreund; Konrad Sandhoff; Richard L. Proia

Gangliosides are a family of glycosphingolipids that contain sialic acid. Although they are abundant on neuronal cell membranes, their precise functions and importance in the central nervous system (CNS) remain largely undefined. We have disrupted the gene encoding GD3 synthase (GD3S), a sialyltransferase expressed in the CNS that is responsible for the synthesis of b-series gangliosides. GD3S−/− mice, even with an absence of b-series gangliosides, appear to undergo normal development and have a normal life span. To further restrict the expression of gangliosides, the GD3S mutant mice were crossbred with mice carrying a disrupted GalNAcT gene encoding β1,4-N-acetylgalactosaminyltransferase. These double mutant mice expressed GM3 as their major ganglioside. In contrast to the single mutant mice, the double mutants displayed a sudden death phenotype and were extremely susceptible to induction of lethal seizures by sound stimulus. These results demonstrate unequivocally that gangliosides play an essential role in the proper functioning of the CNS.


Journal of Biological Chemistry | 1998

Embryonic Stem Cells with a Disrupted GD3 Synthase Gene Undergo Neuronal Differentiation in the Absence of b-Series Gangliosides

Hiromichi Kawai; Kazunori Sango; Katherine A. Mullin; Richard L. Proia

The dramatic changes in the expression of GD3 and other b-series gangliosides during neuronal development and morphogenesis have led to a widely held belief that these gangliosides may be necessary for neuronal differentiation. To determine directly if GD3 and b-series gangliosides are required for neuronal differentiation, we have produced embryonic stem (ES) cells with both alleles of the GD3 synthase gene (GD3S) disrupted by successive rounds of gene targeting. The double-targeted ES cells were deficient in GD3 synthase activity and did not synthesize b-series gangliosides. Despite this deficit, the GD3S(−/−) ES cells could be induced to undergo neuronal differentiation. Neuronally differentiated wild-type and GD3S(−/−) ES cells formed a complex neurite network around the embryoid bodies. Both types of neuronal cells expressed the axon-specific cytoskeletal proteins, neurofilament-M, and growth-associated protein-43 as well as the dendrite-specific marker, microtubule-associated protein-2. Our results indicate that GD3 synthase and b-series gangliosides are not necessary for the neuronal differentiation of uncommitted precursor cells.


Molecular and Cellular Neuroscience | 2010

An in vitro model of the inhibition of axon growth in the lesion scar formed after central nervous system injury.

Junko Kimura-Kuroda; Xichuan Teng; Yukari Komuta; Nozomu Yoshioka; Kazunori Sango; Koki Kawamura; Geoffrey Raisman; Hitoshi Kawano

After central nervous system (CNS) injury, meningeal fibroblasts migrate in the lesion center to form a fibrotic scar which is surrounded by end feet of reactive astrocytes. The fibrotic scar expresses various axonal growth-inhibitory molecules and creates a major impediment for axonal regeneration. We developed an in vitro model of the scar using coculture of cerebral astrocytes and meningeal fibroblasts by adding transforming growth factor-beta1 (TGF-beta1), a potent fibrogenic factor. Addition of TGF-beta1 to this coculture resulted in enhanced proliferation of fibroblasts and the formation of cell clusters which consisted of fibroblasts inside and surrounded by astrocytes. The cell cluster in culture densely accumulated the extracellular matrix molecules and axonal growth-inhibitory molecules similar to the fibrotic scar, and remarkably inhibited the neurite outgrowth of cerebellar neurons. Therefore, this culture system can be available to analyze the inhibitory property in the lesion site of CNS.


Neurochemistry International | 2013

GDNF promotes neurite outgrowth and upregulates galectin-1 through the RET/PI3K signaling in cultured adult rat dorsal root ganglion neurons.

Shizuka Takaku; Hiroko Yanagisawa; Kazuhiko Watabe; Hidenori Horie; Toshihiko Kadoya; Kunihiko Sakumi; Yusaku Nakabeppu; Françoise Poirier; Kazunori Sango

Galectin-1 (GAL-1), a member of a family of β-galactoside binding animal lectins, is predominantly expressed in isolectin B4 (IB4)-binding small non-peptidergic (glial cell line-derived neurotrophic factor (GDNF)-responsive) sensory neurons in the sections of adult rat dorsal root ganglia (DRG), but its functional role and the regulatory mechanisms of its expression in the peripheral nervous system remain unclear. In the present study, both recombinant nerve growth factor (NGF) and GDNF (50 ng/ml) promoted neurite outgrowth from cultured adult rat DRG neurons, whereas GDNF, but not NGF, significantly increased the number of IB4-binding neurons and the relative protein expression of GAL-1 in the neuron-enriched culture of DRG. The GAL-1 expression in immortalized adult rat Schwann cells IFRS1 and DRG neuron-IFRS1 cocultures was unaltered by treatment with GDNF, which suggests that GDNF/GAL-1 signaling axis is more related to neurite outgrowth, rather than neuron-Schwann cell interactions. The GDNF-induced neurite outgrowth and GAL-1 upregulation were attenuated by anti-GDNF family receptor (RET) antibody and phosphatidyl inositol-3-phosphate-kinase (PI3K) inhibitor LY294002, suggesting that the neurite-outgrowth promoting activity of GDNF may be attributable, at least partially, to the upregulation of GAL-1 through RET-PI3K pathway. On the contrary, no significant differences were observed between GAL-1 knockout and wild-type mice in DRG neurite outgrowth in the presence or absence of GDNF. Considerable immunohistochemical colocalization of GAL-3 with GAL-1 in DRG sections and GDNF-induced upregulation of GAL-3 in cultured DRG neurons imply the functional redundancy between these galectins.


Journal of Neuroscience Research | 2011

Spontaneously immortalized Schwann cells from adult Fischer rat as a valuable tool for exploring neuron–Schwann cell interactions

Kazunori Sango; Hiroko Yanagisawa; Emiko Kawakami; Shizuka Takaku; Kyoko Ajiki; Kazuhiko Watabe

We established spontaneously immortalized Schwann cell lines from long‐term cultures of adult Fischer 344 rat dorsal root ganglia (DRG) and peripheral nerves. One of these cell lines, designated immortalized Fischer rat Schwann cells 1 (IFRS1), showed spindle‐shaped morphology; immunoreactivity for S100, p75 neurotrophin receptor (p75NTR), glial fibrillary acidic protein (GFAP), laminin, and vimentin; and mRNA expression of neurotrophic factors (NGF, GDNF, and CNTF), neurotrophin receptors (p75NTR, truncated TrkB, and TrkC), cell adhesion molecules (L1, NCAM, and N‐cadherin), myelin proteins [P0, PMP22, and myelin‐associated glycoprotein (MAG)], transcription factors (Krox20, Sox10, and Oct6), neuregulin‐1 receptors (ErbB2 and ErbB3), and an orphan G protein‐coupled receptor (Gpr126). Conditioned medium (CM) obtained from IFRS1 cells exhibited potent biological activity for the promotion of neuronal survival and neurite outgrowth of cultured adult rat DRG neurons. Furthermore, light and electron microscopic analyses revealed that IFRS1 cells were capable of myelinating neurites while in coculture with adult rat DRG neurons. These findings indicate that IFRS1 cells possess some biological properties of mature Schwann cells and that the coculture system with adult DRG neurons and IFRS1 cells can be a useful tool for the study of peripheral nerve degeneration and regeneration.


Neuroscience Research | 2010

Pleiotrophin induces neurite outgrowth and up-regulates growth-associated protein (GAP)-43 mRNA through the ALK/GSK3β/β-catenin signaling in developing mouse neurons

Hiroko Yanagisawa; Yukari Komuta; Hitoshi Kawano; Masashi Toyoda; Kazunori Sango

Pleiotrophin (PTN) is highly expressed in the nervous system during embryogenesis; however, little is known about its functional role in neural development. By using whole mount in situ hybridization, we observed that the expression pattern of PTN was similar to that of Wnt3a; PTN mRNA was abundant in the nervous tissue along the dorsal midline and in the forelimb and hindlimb buds of embryonic mice (E8.5-E12.5). Treatment with recombinant PTN (100ng/ml) induced phosphorylation of glycogen synthase kinase 3beta (GSK3beta), nuclear localization of beta-catenin and up-regulation of growth-associated protein (GAP)-43 mRNA in cultured embryonic mouse (E14.5) neurons. Furthermore, recombinant PTN enhanced neurite outgrowth from cortical explants embedded in Matrigel. These PTN-induced biochemical changes and neurite outgrowth were attenuated by the co-treatment with anti-anaplastic lymphoma kinase (ALK) antibodies, but not with anti-protein tyrosine phosphatase (PTP)zeta antibodies. These findings imply that ALK is involved in the PTN signaling on neural development.


Histochemistry and Cell Biology | 2012

Myelination in coculture of established neuronal and Schwann cell lines.

Kazunori Sango; Emiko Kawakami; Hiroko Yanagisawa; Shizuka Takaku; Masami Tsukamoto; Kazunori Utsunomiya; Kazuhiko Watabe

Establishing stable coculture systems with neuronal and Schwann cell lines has been considered difficult, presumably because of their high proliferative activity and phenotypic differences from primary cultured cells. The present study is aimed at developing methods for myelin formation under coculture of the neural crest-derived pheochromocytoma cell line PC12 and the immortalized adult rat Schwann cell line IFRS1. Prior to coculture, PC12 cells were seeded at low density (3xa0×xa0102/cm2) and maintained in serum-free medium with N2 supplement, ascorbic acid (50xa0μg/ml), and nerve growth factor (NGF) (50xa0ng/ml) for a week. Exposure to such a NGF-rich environment with minimum nutrients accelerated differentiation and neurite extension, but not proliferation, of PC12 cells. When IFRS1 cells were added to NGF-primed PC12 cells, the cell density ratio of PC12 cells to IFRS1 cells was adjusted from 1:50 to 1:100. The cocultured cells were then maintained in serum-free medium with B27 supplement, ascorbic acid (50xa0μg/ml), NGF (10xa0ng/ml), and recombinant soluble neuregulin-1 type III (25xa0ng/ml). Myelin formation was illustrated by light and electron microscopy performed at day 28 of coculture. The stable PC12-IFRS1 coculture system is free of technical and ethical problems arising from the primary culture and can be a valuable tool to study peripheral nerve degeneration and regeneration.


Experimental Diabetes Research | 2011

Immortalized Adult Rodent Schwann Cells as In Vitro Models to Study Diabetic Neuropathy

Kazunori Sango; Hiroko Yanagisawa; Shizuka Takaku; Emiko Kawakami; Kazuhiko Watabe

We have established spontaneously immortalized Schwann cell lines from normal adult mice and rats and murine disease models. One of the normal mouse cell lines, IMS32, possesses some biological properties of mature Schwann cells and high proliferative activities. The IMS32 cells under hyperglycemic and/or hyperlipidemic conditions have been utilized to investigate the pathogenesis of diabetic neuropathy, especially the polyol pathway hyperactivity, glycation, increased oxidative stress, and reduced synthesis of neurotrophic factors. In addition to the mouse cell lines, our current study focuses on the characterization of a normal rat cell line, IFRS1, under normal and high glucose conditions. These Schwann cell lines can be valuable tools for exploring the detailed mechanisms leading to diabetic neuropathy and novel therapeutic approaches against that condition.


International Journal of Molecular Sciences | 2016

Incretin-Based Therapies for Diabetic Complications: Basic Mechanisms and Clinical Evidence.

Daiji Kawanami; Keiichiro Matoba; Kazunori Sango; Kazunori Utsunomiya

An increase in the rates of morbidity and mortality associated with diabetic complications is a global concern. Glycemic control is important to prevent the development and progression of diabetic complications. Various classes of anti-diabetic agents are currently available, and their pleiotropic effects on diabetic complications have been investigated. Incretin-based therapies such as dipeptidyl peptidase (DPP)-4 inhibitors and glucagon-like peptide-1 receptor agonists (GLP-1RA) are now widely used in the treatment of patients with type 2 diabetes. A series of experimental studies showed that incretin-based therapies have beneficial effects on diabetic complications, independent of their glucose-lowering abilities, which are mediated by anti-inflammatory and anti-oxidative stress properties. Based on these findings, clinical studies to assess the effects of DPP-4 inhibitors and GLP-1RA on diabetic microvascular and macrovascular complications have been performed. Several but not all studies have provided evidence to support the beneficial effects of incretin-based therapies on diabetic complications in patients with type 2 diabetes. We herein discuss the experimental and clinical evidence of incretin-based therapy for diabetic complications.


Neuropathology | 2014

Adenoviral expression of TDP‐43 and FUS genes and shRNAs for protein degradation pathways in rodent motoneurons in vitro and in vivo

Kazuhiko Watabe; Keiko Akiyama; Emiko Kawakami; Tomohiro Ishii; Kentaro Endo; Hiroko Yanagisawa; Kazunori Sango; Masami Tsukamoto

Formation of cytoplasmic aggregates in neuronal and glial cells is one of the pathological hallmarks of amyotrophic lateral sclerosis (ALS). Mutations in two genes encoding transactivation response (TAR) DNA‐binding protein 43 (TDP‐43) and fused in sarcoma (FUS), both of which are main constituents of cytoplasmic aggregates, have been identified in patients with familial and sporadic ALS. Impairment of protein degradation machineries has also been recognized to participate in motoneuron degeneration in ALS. In the present study, we produced recombinant adenovirus vectors encoding wild type and mutant TDP‐43 and FUS, and those encoding short hairpin RNAs (shRNAs) for proteasome (PSMC1), autophagy (ATG5), and endosome (VPS24) systems to investigate whether the coupled gene transductions in motoneurons by these adenoviruses elicit ALS pathology. Cultured neurons, astrocytes and oligodendrocytes differentiated from adult rat neural stem cells and motoneurons derived from mouse embryonic stem cells were successfully infected with these adenoviruses showing cytoplasmic aggregate formation. When these adenoviruses were injected into the facial nerves of adult rats, exogenous TDP‐43 and FUS proteins were strongly expressed in facial motoneurons by a retrograde axonal transport of the adenoviruses. Co‐infections of adenovirus encoding shRNA for PSMC1, ATG5 or VPS24 with TDP‐43 or FUS adenovirus enhanced cytoplasmic aggregate formation in facial motoneurons, suggesting that impairment of protein degradation pathways accelerates formation of TDP‐43 and FUS‐positive aggregates in ALS.

Collaboration


Dive into the Kazunori Sango's collaboration.

Top Co-Authors

Avatar

Kazuhiko Watabe

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hiroko Yanagisawa

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Shizuka Takaku

Tokyo Medical University

View shared research outputs
Top Co-Authors

Avatar

Kazunori Utsunomiya

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Masami Tsukamoto

Jikei University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Richard L. Proia

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Hidenori Horie

Yokohama City University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Junji Yamauchi

Tokyo University of Pharmacy and Life Sciences

View shared research outputs
Top Co-Authors

Avatar

Toshihiko Kadoya

Maebashi Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge