Network


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

Hotspot


Dive into the research topics where Tsukasa Sanosaka is active.

Publication


Featured researches published by Tsukasa Sanosaka.


Developmental Cell | 2009

Committed Neuronal Precursors Confer Astrocytic Potential on Residual Neural Precursor Cells

Masakazu Namihira; Jun Kohyama; Katsunori Semi; Tsukasa Sanosaka; Benjamin Deneen; Tetsuya Taga; Kinichi Nakashima

During midgestation, mammalian neural precursor cells (NPCs) differentiate only into neurons. Generation of astrocytes is prevented at this stage, because astrocyte-specific gene promoters are methylated. How the subsequent switch from suppression to expression of astrocytic genes occurs is unknown. We show in this study that Notch ligands are expressed on committed neuronal precursors and young neurons in mid-gestational telencephalon, and that neighboring Notch-activated NPCs acquire the potential to become astrocytes. Activation of the Notch signaling pathway in midgestational NPCs induces expression of the transcription factor nuclear factor I, which binds to astrocytic gene promoters, resulting in demethylation of astrocyte-specific genes. These findings provide a mechanistic explanation for why neurons come first: committed neuronal precursors and young neurons potentiate remaining NPCs to differentiate into the next cell lineage, astrocytes.


Stem Cells | 2012

Treatment of a Mouse Model of Spinal Cord Injury by Transplantation of Human Induced Pluripotent Stem Cell-Derived Long-Term Self-Renewing Neuroepithelial-Like Stem Cells†‡§

Yusuke Fujimoto; Masahiko Abematsu; Keita Tsujimura; Tsukasa Sanosaka; Berry Juliandi; Katsunori Semi; Masakazu Namihira; Setsuro Komiya; Austin Smith; Kinichi Nakashima

Because of their ability to self‐renew, to differentiate into multiple lineages, and to migrate toward a damaged site, neural stem cells (NSCs), which can be derived from various sources such as fetal tissues and embryonic stem cells, are currently considered to be promising components of cell replacement strategies aimed at treating injuries of the central nervous system, including the spinal cord. Despite their efficiency in promoting functional recovery, these NSCs are not homogeneous and possess variable characteristics depending on their derivation protocols. The advent of induced pluripotent stem (iPS) cells has provided new prospects for regenerative medicine. We used a recently developed robust and stable protocol for the generation of long‐term, self‐renewing, neuroepithelial‐like stem cells from human iPS cells (hiPS‐lt‐NES cells), which can provide a homogeneous and well‐defined population of NSCs for standardized analysis. Here, we show that transplanted hiPS‐lt‐NES cells differentiate into neural lineages in the mouse model of spinal cord injury (SCI) and promote functional recovery of hind limb motor function. Furthermore, using two different neuronal tracers and ablation of the transplanted cells, we revealed that transplanted hiPS‐lt‐NES cell‐derived neurons, together with the surviving endogenous neurons, contributed to restored motor function. Both types of neurons reconstructed the corticospinal tract by forming synaptic connections and integrating neuronal circuits. Our findings indicate that hiPS‐lt‐NES transplantation represents a promising avenue for effective cell‐based treatment of SCI. STEM CELLS2012;30:1163–1173


Stem Cells | 2009

Astrocyte Differentiation of Neural Precursor Cells is Enhanced by Retinoic Acid Through a Change in Epigenetic Modification

Hirotsugu Asano; Makoto Aonuma; Tsukasa Sanosaka; Jun Kohyama; Masakazu Namihira; Kinichi Nakashima

Neurons, astrocytes, and oligodendrocytes—the three major cell types that comprise the central nervous system—are generated from common multipotent neural precursor cells (NPCs). Members of the interleukin‐6 family of cytokines, including leukemia inhibitory factor (LIF), induce astrocyte differentiation of NPCs by activating the transcription factor signal transducer and activator of transcription 3 (STAT3). We show here that retinoic acid (RA) facilitates LIF‐induced astrocyte differentiation of NPCs. RA and LIF synergistically activate the promoter of gfap, which encodes the astrocytic marker glial fibrillary acidic protein, and a putative RA response element in the promoter was found to be critical for this activation. Histone H3 acetylation around the STAT‐binding site in the gfap promoter was increased in NPCs treated with RA, allowing STAT3 to gain access to the promoter more efficiently. These results suggest that RA acts in concert with LIF to induce astrocyte differentiation of NPCs through an epigenetic mechanism that involves cross‐talk between distinct signaling pathways. Stem Cells 2009;27:2744–2752


Embo Molecular Medicine | 2011

Insulin biosynthesis in neuronal progenitors derived from adult hippocampus and the olfactory bulb

Tomoko Kuwabara; Mohamedi N. Kagalwala; Yasuko Onuma; Yuzuru Ito; Masaki Warashina; Kazuyuki Terashima; Tsukasa Sanosaka; Kinichi Nakashima; Fred H. Gage; Makoto Asashima

In the present study, we demonstrated that insulin is produced not only in the mammalian pancreas but also in adult neuronal cells derived from the hippocampus and olfactory bulb (OB). Paracrine Wnt3 plays an essential role in promoting the active expression of insulin in both hippocampal and OB‐derived neural stem cells. Our analysis indicated that the balance between Wnt3, which triggers the expression of insulin via NeuroD1, and IGFBP‐4, which inhibits the original Wnt3 action, is regulated depending on diabetic (DB) status. We also show that adult neural progenitors derived from DB animals retain the ability to give rise to insulin‐producing cells and that grafting neuronal progenitors into the pancreas of DB animals reduces glucose levels. This study provides an example of a simple and direct use of adult stem cells from one organ to another, without introducing additional inductive genes.


Journal of Cell Biology | 2010

BMP-induced REST regulates the establishment and maintenance of astrocytic identity

Jun Kohyama; Tsukasa Sanosaka; Akinori Tokunaga; Eriko Takatsuka; Keita Tsujimura; Hideyuki Okano; Kinichi Nakashima

Astrocyte differentiation and maintenance is promoted by BMP signaling, which induces REST/NRSF to repress neuronal genes.


Journal of Neuroscience Research | 2006

Methyl-CpG binding proteins are involved in restricting differentiation plasticity in neurons.

Hiroki Setoguchi; Masakazu Namihira; Jun Kohyama; Hirotsugu Asano; Tsukasa Sanosaka; Kinichi Nakashima

Neurons and astrocytes are generated from common neural precursors, yet neurogenesis precedes astrocytogenesis, which normally commences at later stages of development. We have previously reported that a particular cytosine residue within a STAT3‐binding site in the astrocyte‐specific marker glial fibrillary acidic protein (GFAP) gene promoter becomes demethylated in neuroepithelial cells as gestation proceeds. This demethylation correlates tightly with the onset of astrocyte differentiation, suggesting that a change in DNA methylation at cell‐type‐specific gene promoters controls the switch from neurogenesis to astrocytogenesis in the developing brain. Here, we show that late‐gestation neuroepithelial cells, which have already lost the methylation in the STAT3‐binding site within the GFAP promoter, can still give rise to neurons and that these neurons do not respond to a STAT3‐activating cytokine to express GFAP. Members of a transcriptional repressor family, the methylated‐CpG binding proteins (MBDs), including MeCP2, are predominantly expressed in neurons, and ectopic MeCP2 expression inhibited astrocyte differentiation of neuroepithelial cells. Moreover, we found that exon 1 of the GFAP gene remains hypermethylated even in neuroepithelial cells at a late developmental stage and in neurons differentiated from such neuroepithelial cells. We further demonstrate that MeCP2 actually binds to the highly methylated exon 1 of the GFAP gene in neurons. These results suggest that region‐specific DNA methylation and MBDs play an important role in the regulation of differentiation plasticity in neurons.


Molecular Brain | 2015

Differentiation of multipotent neural stem cells derived from Rett syndrome patients is biased toward the astrocytic lineage

Tomoko Andoh-Noda; Wado Akamatsu; Kunio Miyake; Takuya Matsumoto; Ryo Yamaguchi; Tsukasa Sanosaka; Yohei Okada; Tetsuro Kobayashi; Manabu Ohyama; Kinichi Nakashima; Hiroshi Kurosawa; Takeo Kubota; Hideyuki Okano

BackgroundRett syndrome (RTT) is one of the most prevalent neurodevelopmental disorders in females, caused by de novo mutations in the X-linked methyl CpG-binding protein 2 gene, MECP2. Although abnormal regulation of neuronal genes due to mutant MeCP2 is thought to induce autistic behavior and impaired development in RTT patients, precise cellular mechanisms underlying the aberrant neural progression remain unclear.ResultsTwo sets of isogenic pairs of either wild-type or mutant MECP2-expressing human induced pluripotent stem cell (hiPSC) lines were generated from a single pair of 10-year-old RTT-monozygotic (MZ) female twins. Mutant MeCP2-expressing hiPSC lines did not express detectable MeCP2 protein during any stage of differentiation. The lack of MeCP2 reflected altered gene expression patterns in differentiated neural cells rather than in undifferentiated hiPSCs, as assessed by microarray analysis. Furthermore, MeCP2 deficiency in the neural cell lineage increased astrocyte-specific differentiation from multipotent neural stem cells. Additionally, chromatin immunoprecipitation (ChIP) and bisulfite sequencing assays indicated that anomalous glial fibrillary acidic protein gene (GFAP) expression in the MeCP2-negative, differentiated neural cells resulted from the absence of MeCP2 binding to the GFAP gene.ConclusionsAn isogenic RTT-hiPSC model demonstrated that MeCP2 participates in the differentiation of neural cells. Moreover, MeCP2 deficiency triggers perturbation of astrocytic gene expression, yielding accelerated astrocyte formation from RTT-hiPSC-derived neural stem cells. These findings are likely to shed new light on astrocytic abnormalities in RTT, and suggest that astrocytes, which are required for neuronal homeostasis and function, might be a new target of RTT therapy.


Nature Communications | 2012

Unfolded protein response, activated by OASIS family transcription factors, promotes astrocyte differentiation

Atsushi Saito; Soshi Kanemoto; Noritaka Kawasaki; Rie Asada; Hideo Iwamoto; Mami Oki; Hidetaka Miyagi; Soutarou Izumi; Tsukasa Sanosaka; Kinichi Nakashima; Kazunori Imaizumi

OASIS is a member of the CREB/ATF family of transcription factors and modulates cell- or tissue-specific unfolded protein response signalling. Here we show that this modulation has a critical role in the differentiation of neural precursor cells into astrocytes. Cerebral cortices of mice specifically deficient in OASIS (Oasis(-/-)) contain fewer astrocytes and more neural precursor cells than those of wild-type mice during embryonic development. Furthermore, astrocyte differentiation is delayed in primary cultured Oasis(-/-) neural precursor cells. The transcription factor Gcm1, which is necessary for astrocyte differentiation in Drosophila, is revealed to be a target of OASIS. Introduction of Gcm1 into Oasis(-/-) neural precursor cells improves the delayed differentiation of neural precursor cells into astrocytes by accelerating demethylation of the Gfap promoter. Gcm1 expression is temporally controlled by the unfolded protein response through interactions between OASIS family members during astrocyte differentiation. Taken together, our findings demonstrate a novel mechanism by which OASIS and its associated family members are modulated by the unfolded protein response to finely control astrocyte differentiation.


Neuroscience | 2008

Identification of genes that restrict astrocyte differentiation of midgestational neural precursor cells.

Tsukasa Sanosaka; Masakazu Namihira; Hirotsugu Asano; Jun Kohyama; K. Aisaki; K. Igarashi; Jun Kanno; Kinichi Nakashima

During development of the mammalian CNS, neurons and glial cells (astrocytes and oligodendrocytes) are generated from common neural precursor cells (NPCs). However, neurogenesis precedes gliogenesis, which normally commences at later stages of fetal telencephalic development. Astrocyte differentiation of mouse NPCs at embryonic day (E) 14.5 (relatively late gestation) is induced by activation of the transcription factor signal transducer and activator of transcription (STAT) 3, whereas at E11.5 (mid-gestation) NPCs do not differentiate into astrocytes even when stimulated by STAT3-activating cytokines such as leukemia inhibitory factor (LIF). This can be explained in part by the fact that astrocyte-specific gene promoters are highly methylated in NPCs at E11.5, but other mechanisms are also likely to play a role. We therefore sought to identify genes involved in the inhibition of astrocyte differentiation of NPCs at midgestation. We first examined gene expression profiles in E11.5 and E14.5 NPCs, using Affymetrix GeneChip analysis, applying the Percellome method to normalize gene expression level. We then conducted in situ hybridization analysis for selected genes found to be highly expressed in NPCs at midgestation. Among these genes, we found that N-myc and high mobility group AT-hook 2 (Hmga2) were highly expressed in the E11.5 but not the E14.5 ventricular zone of mouse brain, where NPCs reside. Transduction of N-myc and Hmga2 by retroviruses into E14.5 NPCs, which normally differentiate into astrocytes in response to LIF, resulted in suppression of astrocyte differentiation. However, sustained expression of N-myc and Hmga2 in E11.5 NPCs failed to maintain the hypermethylated status of an astrocyte-specific gene promoter. Taken together, our data suggest that astrocyte differentiation of NPCs is regulated not only by DNA methylation but also by genes whose expression is controlled spatio-temporally during brain development.


Stem Cells | 2012

Oxygen levels epigenetically regulate fate switching of neural precursor cells via hypoxia-inducible factor 1α-Notch signal interaction in the developing brain

Tetsuji Mutoh; Tsukasa Sanosaka; Kei Ito; Kinichi Nakashima

Oxygen levels in tissues including the embryonic brain are lower than those in the atmosphere. We reported previously that Notch signal activation induces demethylation of astrocytic genes, conferring astrocyte differentiation ability on midgestational neural precursor cells (mgNPCs). Here, we show that the oxygen sensor hypoxia‐inducible factor 1α (HIF1α) plays a critical role in astrocytic gene demethylation in mgNPCs by cooperating with the Notch signaling pathway. Expression of constitutively active HIF1α and a hyperoxic environment, respectively, promoted and impeded astrocyte differentiation in the developing brain. Our findings suggest that hypoxia contributes to the appropriate scheduling of mgNPC fate determination. STEM CELLS 2012;30:561–569

Collaboration


Dive into the Tsukasa Sanosaka's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jun Kohyama

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Masakazu Namihira

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Keita Tsujimura

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar

Berry Juliandi

Bogor Agricultural University

View shared research outputs
Top Co-Authors

Avatar

Katsunori Semi

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hirotsugu Asano

Nara Institute of Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge