Network


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

Hotspot


Dive into the research topics where Saya Ito is active.

Publication


Featured researches published by Saya Ito.


Neuron | 2002

Androgen-dependent neurodegeneration by polyglutamine-expanded human androgen receptor in Drosophila.

Ken-ichi Takeyama; Saya Ito; Ayako Yamamoto; Hiromu Tanimoto; Takashi Furutani; Hirotaka Kanuka; Masayuki Miura; Tetsuya Tabata; Shigeaki Kato

Spinal and bulbar muscular atrophy (SBMA) is an X-linked, adult-onset, neurodegenerative disorder affecting only males and is caused by expanded polyglutamine (polyQ) stretches in the N-terminal A/B domain of human androgen receptor (hAR). Although no overt phenotype was detected in adult fly eye photoreceptor neurons expressing mutant hAR (polyQ 52), ingestion of androgen or its known antagonists caused marked neurodegeneration with nuclear localization and structural alteration of the hAR mutant. Ligand-independent toxicity was detected with a truncated polyQ-expanded A/B domain alone, which was attenuated with cytosolic trapping by coexpression of the unliganded hAR E/F ligand binding domain. Thus, our findings suggest that the full binding of androgen to the polyQ-expanded hAR mutants leads to structural alteration with nuclear translocation that eventually results in the onset of SBMA in male patients.


Molecular Cell | 2008

A TFTC/STAGA Module Mediates Histone H2A and H2B Deubiquitination, Coactivates Nuclear Receptors, and Counteracts Heterochromatin Silencing

Yue Zhao; Guillaume Lang; Saya Ito; Jacques Bonnet; Eric Metzger; Shun Sawatsubashi; Eriko Suzuki; Xavier Le Guezennec; Hendrik G. Stunnenberg; Aleksey N. Krasnov; S. G. Georgieva; Roland Schüle; Ken-ichi Takeyama; Shigeaki Kato; Laszlo Tora; Didier Devys

Transcriptional activators, several different coactivators, and general transcription factors are necessary to access specific loci in the dense chromatin structure to allow precise initiation of RNA polymerase II (Pol II) transcription. Histone acetyltransferase (HAT) complexes were implicated in loosening the chromatin around promoters and thus in gene activation. Here we demonstrate that the 2 MDa GCN5 HAT-containing metazoan TFTC/STAGA complexes contain a histone H2A and H2B deubiquitinase activity. We have identified three additional subunits of TFTC/STAGA (ATXN7L3, USP22, and ENY2) that form the deubiquitination module. Importantly, we found that this module is an enhancer of position effect variegation in Drosophila. Furthermore, we demonstrate that ATXN7L3, USP22, and ENY2 are required as cofactors for the full transcriptional activity by nuclear receptors. Thus, the deubiquitinase activity of the TFTC/STAGA HAT complex is necessary to counteract heterochromatin silencing and acts as a positive cofactor for activation by nuclear receptors in vivo.


Nature | 2011

GlcNAcylation of histone H2B facilitates its monoubiquitination

Ryoji Fujiki; Waka Hashiba; Hiroki Sekine; Atsushi Yokoyama; Toshihiro Chikanishi; Saya Ito; Yuuki Imai; Jae-Hoon Kim; Housheng Hansen He; Katsuhide Igarashi; Jun Kanno; Fumiaki Ohtake; Hirochika Kitagawa; Robert G. Roeder; Myles Brown; Shigeaki Kato

Chromatin reorganization is governed by multiple post-translational modifications of chromosomal proteins and DNA. These histone modifications are reversible, dynamic events that can regulate DNA-driven cellular processes. However, the molecular mechanisms that coordinate histone modification patterns remain largely unknown. In metazoans, reversible protein modification by O-linked N-acetylglucosamine (GlcNAc) is catalysed by two enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). However, the significance of GlcNAcylation in chromatin reorganization remains elusive. Here we report that histone H2B is GlcNAcylated at residue S112 by OGT in vitro and in living cells. Histone GlcNAcylation fluctuated in response to extracellular glucose through the hexosamine biosynthesis pathway (HBP). H2B S112 GlcNAcylation promotes K120 monoubiquitination, in which the GlcNAc moiety can serve as an anchor for a histone H2B ubiquitin ligase. H2B S112 GlcNAc was localized to euchromatic areas on fly polytene chromosomes. In a genome-wide analysis, H2B S112 GlcNAcylation sites were observed widely distributed over chromosomes including transcribed gene loci, with some sites co-localizing with H2B K120 monoubiquitination. These findings suggest that H2B S112 GlcNAcylation is a histone modification that facilitates H2BK120 monoubiquitination, presumably for transcriptional activation.


Genes & Development | 2010

A histone chaperone, DEK, transcriptionally coactivates a nuclear receptor

Shun Sawatsubashi; Takuya Murata; Jinseon Lim; Ryoji Fujiki; Saya Ito; Eriko Suzuki; Masahiko Tanabe; Yue Zhao; Shuhei Kimura; Sally Fujiyama; Takashi Ueda; Daiki Umetsu; Takashi Ito; Ken-ichi Takeyama; Shigeaki Kato

Chromatin reorganization is essential for transcriptional control by sequence-specific transcription factors. However, the molecular link between transcriptional control and chromatin reconfiguration remains unclear. By colocalization of the nuclear ecdysone receptor (EcR) on the ecdysone-induced puff in the salivary gland, Drosophila DEK (dDEK) was genetically identified as a coactivator of EcR in both insect cells and intact flies. Biochemical purification and characterization of the complexes containing fly and human DEKs revealed that DEKs serve as histone chaperones via phosphorylation by forming complexes with casein kinase 2. Consistent with the preferential association of the DEK complex with histones enriched in active epigenetic marks, dDEK facilitated H3.3 assembly during puff formation. In some human myeloid leukemia patients, DEK was fused to CAN by chromosomal translocation. This mutation significantly reduced formation of the DEK complex, which is required for histone chaperone activity. Thus, the present study suggests that at least one histone chaperone can be categorized as a type of transcriptional coactivator for nuclear receptors.


Molecular and Cellular Biology | 2007

The Pituitary Function of Androgen Receptor Constitutes a Glucocorticoid Production Circuit

Junko Miyamoto; Takahiro Matsumoto; Hiroko Shiina; Kazuki Inoue; Ichiro Takada; Saya Ito; Johbu Itoh; Takeo Minematsu; Takashi Sato; Toshihiko Yanase; Hajime Nawata; Yoshiyuki Osamura; Shigeaki Kato

ABSTRACT Androgen receptor (AR) mediates diverse androgen actions, particularly reproductive processes in males and females. AR-mediated androgen signaling is considered to also control metabolic processes; however, the molecular basis remains elusive. In the present study, we explored the molecular mechanism of late-onset obesity in male AR null mutant (ARKO) mice. We determined that the obesity was caused by a hypercorticoid state. The negative feedback system regulating glucocorticoid production was impaired in ARKO mice. Male and female ARKO mice exhibited hypertrophic adrenal glands and glucocorticoid overproduction, presumably due to high levels of adrenal corticotropic hormone. The pituitary glands of the ARKO males had increased expression of proopiomelanocortin and decreased expression of the glucocorticoid receptor (GR). There were no overt structural abnormalities and no alteration in the distribution of cell types in the pituitaries of male ARKO mice. Additionally, there was normal production of the other hormones within the glucocorticoid feedback system in both the pituitary and hypothalamus. In a cell line derived from pituitary glands, GR expression was under the positive control of the activated AR. Thus, this study suggests that the activated AR supports the negative feedback regulation of glucocorticoid production via up-regulation of GR expression in the pituitary gland.


Molecular and Cellular Biology | 2009

Corepressive Action of CBP on Androgen Receptor Transactivation in Pericentric Heterochromatin in a Drosophila Experimental Model System

Yue Zhao; Ken-ichi Takeyama; Shun Sawatsubashi; Saya Ito; Eriko Suzuki; Kaoru Yamagata; Masahiko Tanabe; Shuhei Kimura; Sally Fujiyama; Takashi Ueda; Takuya Murata; Hiroyuki Matsukawa; Yuko Shirode; Alexander Kouzmenko; Feng Li; T. Tabata; Shigeaki Kato

ABSTRACT Ligand-bound nuclear receptors (NR) activate transcription of the target genes. This activation is coupled with histone modifications and chromatin remodeling through the function of various coregulators. However, the nature of the dependence of a NR coregulator action on the presence of the chromatin environment at the target genes is unclear. To address this issue, we have developed a modified position effect variegation experimental model system that includes an androgen-dependent reporter transgene inserted into either a pericentric heterochromatin region or a euchromatic region of Drosophila chromosome. Human androgen receptor (AR) and its constitutively active truncation mutant (AR AF-1) were transcriptionally functional in both chromosomal regions. Predictably, the level of AR-induced transactivation was lower in the pericentric heterochromatin. In genetic screening for AR AF-1 coregulators, Drosophila CREB binding protein (dCBP) was found to corepress AR transactivation at the pericentric region whereas it led to coactivation in the euchromatic area. Mutations of Sir2 acetylation sites or deletion of the CBP acetyltransferase domain abrogated dCBP corepressive action for AR at heterochromatic areas in vivo. Such a CBP corepressor function for AR was observed in the transcriptionally silent promoter of an AR target gene in cultured mammalian cells. Thus, our findings suggest that the action of NR coregulators may depend on the state of chromatin at the target loci.


Oncogene | 2007

Id2 gene-targeted crosstalk between Wnt and retinoid signaling regulates proliferation in human keratinocytes.

A Memezawa; Ichiro Takada; Ken-ichi Takeyama; Mamoru Igarashi; Saya Ito; Setsuya Aiba; Shigeaki Kato; Alexander Kouzmenko

We investigated the effect of all-trans-retinoic acid (atRA) on proliferation in several human skin cell lines and found that antiproliferative potency of atRA correlated with the endogenous activity of canonical Wnt signaling. In HaCaT keratinocytes, we found that atRA significantly suppressed the expression of Id2, a member of the inhibitor of differentiation family of transcription factors that regulate cell growth and differentiation. However, no apparent change in the expression of other Wnt targets, like c-Myc or cyclin D1, was observed. Retinoid-induced Id2 gene suppression was associated with decreased levels of histone H3 and H4 acetylation and histone H3 Lys-4 methylation, and with recruitment of the LSD1 demethylase at the Wnt-response element (WRE) (TCF/LEF-binding site), in the Id2 gene promoter. None of such changes was detected at the WRE of c-Myc and cyclin D1 gene promoters. Inhibition of Id2 by short interfering RNA (siRNA) had a similar effect on the proliferation of HaCaT cells as exposure to atRA, whereas anti-β-catenin siRNA significantly inhibited its antiproliferative effect. These data suggest that downregulation of Id2 gene expression through transcriptional convergence between Wnt and retinoid signaling pathways underlies the antiproliferative effect of retinoids in keratinocytes, and provide evidence of gene-targeted crosstalk between signaling pathways.


Genes to Cells | 2004

In vivo potentiation of human oestrogen receptor alpha by Cdk7-mediated phosphorylation.

Saya Ito; Ken-ichi Takeyama; Ayako Yamamoto; Shun Sawatsubashi; Yuko Shirode; Alexander Kouzmenko; Tetsuya Tabata; Shigeaki Kato

Phosphorylation of the Ser118 residue in the N‐terminal A/B domain of the human oestrogen receptor α (hERα) by mitogen‐activated protein kinase (MAPK), stimulated via growth factor signalling pathways, is known to potentiate ERα ligand‐induced transactivation function. Besides MAPK, cyclin dependent kinase 7 (Cdk7) in the TFIIH complex has also been found to potentiate hERα transactivation in vitro through Ser118 phosphorylation. To investigate an impact of Cdk7 on hERα transactivation in vivo, we assessed activity of hERα in a wild‐type and cdk7 inactive mutant Drosophila that ectopically expressed hERα in the eye disc. Ectopic expression of the wild‐type or mutant receptors, together with a green fluorescent protein (GFP) reporter gene, allowed us to demonstrate that hERα expressed in the fly tissues was transcriptionally functional and adequately responded to hERα ligands in the patterns similar to those observed in mammalian cells. Replacement of Ser118 with alanine in hERα (S118A mutant) significantly reduced the ligand‐induced hERα transactivation function. Importantly, while in cdk7 inactive mutant Drosophila the wild‐type hERα exhibited reduced response to the ligand; levels of transactivation by the hERα S118A mutant were not affected in these inactive cdk7 mutant flies. Furthermore, phosphorylation of hERα at Ser118 has been observed in vitro by both human and Drosophila Cdk7. Our findings demonstrate that Cdk7 is involved in regulation of the ligand‐induced transactivation function of hERαin vivo via Ser118 phosphorylation.


Biochemical and Biophysical Research Communications | 2008

Drosophila arginine methyltransferase 1 (DART1) is an ecdysone receptor co-repressor.

Shuhei Kimura; Shun Sawatsubashi; Saya Ito; Alexander Kouzmenko; Eriko Suzuki; Yue Zhao; Kaoru Yamagata; Masahiko Tanabe; Takashi Ueda; Sari Fujiyama; Takuya Murata; Hiroyuki Matsukawa; Ken-ichi Takeyama; Nobuo Yaegashi; Shigeaki Kato

Histone arginine methylation is an epigenetic marker that regulates gene expression by defining the chromatin state. Arginine methyltransferases, therefore, serve as transcriptional co-regulators. However, unlike other transcriptional co-regulators, the physiological roles of arginine methyltransferases are poorly understood. Drosophila arginine methyltransferase 1 (DART1), the mammalian PRMT1 homologue, methylates the arginine residue of histone H4 (H4R3me2). Disruption of DART1 in Drosophila by imprecise P-element excision resulted in low viability during metamorphosis in the pupal stages. In the pupal stage, an ecdysone hormone signal is critical for developmental progression. DART1 interacted with the nuclear ecdysone receptor (EcR) in a ligand-dependent manner, and co-repressed EcR in intact flies. These findings suggest that DART1, a histone arginine methyltransferase, is a co-repressor of EcR that is indispensable for normal pupal development in the intact fly.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Aberrant E2F activation by polyglutamine expansion of androgen receptor in SBMA neurotoxicity.

Eriko Suzuki; Yue Zhao; Saya Ito; Shun Sawatsubashi; Takuya Murata; Takashi Furutani; Yuko Shirode; Kaoru Yamagata; Masahiko Tanabe; Shuhei Kimura; Takashi Ueda; Sally Fujiyama; Jinseon Lim; Hiroyuki Matsukawa; Alexander Kouzmenko; Toshiro Aigaki; Tetsuya Tabata; Ken-ichi Takeyama; Shigeaki Kato

Spinal and bulbar muscular atrophy (SBMA) is a neurodegenerative disorder caused by a polyglutamine repeat (polyQ) expansion within the human androgen receptor (AR). Unlike other neurodegenerative diseases caused by abnormal polyQ expansion, the onset of SBMA depends on androgen binding to mutant human polyQ-AR proteins. This is also observed in Drosophila eyes ectopically expressing the polyQ-AR mutants. We have genetically screened mediators of androgen-induced neurodegeneration caused by polyQ-AR mutants in Drosophila eyes. We identified Rbf (Retinoblastoma-family protein), the Drosophila homologue of human Rb (Retinoblastoma protein), as a neuroprotective factor. Androgen-dependent association of Rbf or Rb with AR was remarkably potentiated by aberrant polyQ expansion. Such potentiated Rb association appeared to attenuate recruitment of histone deacetyltransferase 1 (HDAC1), a corepressor of E2F function. Either overexpression of Rbf or E2F deficiency in fly eyes reduced the neurotoxicity of the polyQ-AR mutants. Induction of E2F function by polyQ-AR-bound androgen was suppressed by Rb in human neuroblastoma cells. We conclude that abnormal expansion of polyQ may potentiate innate androgen-dependent association of AR with Rb. This appears to lead to androgen-dependent onset of SBMA through aberrant E2F transactivation caused by suppressed histone deacetylation.

Collaboration


Dive into the Saya Ito'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

Masahiko Tanabe

Japanese Foundation for Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge