Network


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

Hotspot


Dive into the research topics where Yuko Saeki is active.

Publication


Featured researches published by Yuko Saeki.


Cell | 2010

Ligand-Specific c-Fos Expression Emerges from the Spatiotemporal Control of ErbB Network Dynamics

Takashi Nakakuki; Marc R. Birtwistle; Yuko Saeki; Noriko Yumoto; Kaori Ide; Takeshi Nagashima; Lutz Brusch; Babatunde A. Ogunnaike; Mariko Okada-Hatakeyama; Boris N. Kholodenko

Activation of ErbB receptors by epidermal growth factor (EGF) or heregulin (HRG) determines distinct cell-fate decisions, although signals propagate through shared pathways. Using mathematical modeling and experimental approaches, we unravel how HRG and EGF generate distinct, all-or-none responses of the phosphorylated transcription factor c-Fos. In the cytosol, EGF induces transient and HRG induces sustained ERK activation. In the nucleus, however, ERK activity and c-fos mRNA expression are transient for both ligands. Knockdown of dual-specificity phosphatases extends HRG-stimulated nuclear ERK activation, but not c-fos mRNA expression, implying the existence of a HRG-induced repressor of c-fos transcription. Further experiments confirmed that this repressor is mainly induced by HRG, but not EGF, and requires new protein synthesis. We show how a spatially distributed, signaling-transcription cascade robustly discriminates between transient and sustained ERK activities at the c-Fos system level. The proposed control mechanisms are general and operate in different cell types, stimulated by various ligands.


BMC Genomics | 2009

Ligand-specific sequential regulation of transcription factors for differentiation of MCF-7 cells

Yuko Saeki; Takaho A. Endo; Kaori Ide; Takeshi Nagashima; Noriko Yumoto; Tetsuro Toyoda; Harukazu Suzuki; Yoshihide Hayashizaki; Yoshiyuki Sakaki; Mariko Okada-Hatakeyama

BackgroundSharing a common ErbB/HER receptor signaling pathway, heregulin (HRG) induces differentiation of MCF-7 human breast cancer cells while epidermal growth factor (EGF) elicits proliferation. Although cell fates resulting from action of the aforementioned ligands completely different, the respective gene expression profiles in early transcription are qualitatively similar, suggesting that gene expression during late transcription, but not early transcription, may reflect ligand specificity. In this study, based on both the data from time-course quantitative real-time PCR on over 2,000 human transcription factors and microarray of all human genes, we identified a series of transcription factors which may control HRG-specific late transcription in MCF-7 cells.ResultsWe predicted that four transcription factors including EGR4, FRA-1, FHL2, and DIPA should have responsibility of regulation in MCF-7 cell differentiation. Validation analysis suggested that one member of the activator protein 1 (AP-1) family, FOSL-1 (FRA-1 gene), appeared immediately following c-FOS expression, might be responsible for expression of transcription factor FHL2 through activation of the AP-1 complex. Furthermore, RNAi gene silencing of FOSL-1 and FHL2 resulted in increase of extracellular signal-regulated kinase (ERK) phosphorylation of which duration was sustained by HRG stimulation.ConclusionOur analysis indicated that a time-dependent transcriptional regulatory network including c-FOS, FRA-1, and FHL2 is vital in controlling the ERK signaling pathway through a negative feedback loop for MCF-7 cell differentiation.


FEBS Journal | 2009

Mutation of epidermal growth factor receptor is associated with MIG6 expression

Takeshi Nagashima; Ryoko Ushikoshi-Nakayama; Kaori Ide; Noriko Yumoto; Yoshimi Naruo; Kaoru Takahashi; Yuko Saeki; Makoto Taiji; Hiroshi Tanaka; Shih-Feng Tsai; Mariko Hatakeyama

Controlled activation of epidermal growth factor receptor (EGFR) is systematically guaranteed at the molecular level; however, aberrant activation of EGFR is frequently found in cancer. Transcription induced by EGFR activation often involves the coordinated expression of genes that positively and negatively regulate the original signaling pathway; therefore, alterations in EGFR kinase activity may reflect changes in gene expression associated with the pathway. In the present study, we investigated transcriptional changes after EGF stimulation with or without the EGFR kinase inhibitor Iressa in H1299 human non‐small‐cell lung cancer cells [parental H1299, H1299 cells that overexpress wild‐type EGFR (EGFR‐WT) and mutant H1299 cells that overexpress EGFR where Leu858 is substituted with Arg (L858R)]. The results obtained clearly demonstrate differences in transcriptional activity in the absence or presence of EGFR kinase activity, with genes sharing the same molecular functions showing distinct expression dynamics. The results show the particular enrichment of EGFR/ErbB signaling‐related genes in a differentially expressed gene set, and significant protein expression of MIG6/RALT(ERRFI1), an EGFR negative regulator, was confirmed in L858R. High MIG6 protein expression was correlated with basal EGFR phosphorylation and inversely correlated with EGF‐induced extracellular signal‐regulated protein kinase phosphorylation levels. Investigation of the NCI‐60 cell lines showed that ERRFI1 expression was correlated with EGFR expression, regardless of tissue type. These results suggest that cells accumulate MIG6 as an inherent negative regulator to suppress excess EGFR activity when basal EGFR kinase activity is considerably high. Taking all the above together, an EGFR mutation can cause transcriptional changes to accommodate the activation potency of the original signaling pathway at the cellular level.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Dynamically varying interactions between heregulin and ErbB proteins detected by single-molecule analysis in living cells

Michio Hiroshima; Yuko Saeki; Mariko Okada-Hatakeyama; Yasushi Sako

Heregulin (HRG) belongs to the family of EGFs and activates the receptor proteins ErbB3 and ErbB4 in a variety of cell types to regulate cell fate. The interactions between HRG and ErbB3/B4 are important to the pathological mechanisms underlying schizophrenia and some cancers. Here, we observed the reaction kinetics between fluorescently labeled single HRG molecules and ErbB3/B4 on the surfaces of MCF-7 human breast cancer cells. The equilibrium association and the dissociation from equilibrium were also measured using single-molecule imaging techniques. The unitary association processes mirrored the EGF and ErbB1 interactions in HeLa cells [Teramura Y, et al. (2006) EMBO J 25:4215–4222], suggesting that the predimerization of the receptors, followed by intermediate formation (between the first and second ligand-binding events to a receptor dimer), accelerated the formation of doubly liganded signaling dimers of the receptor molecules. However, the dissociation analysis suggested that the first HRG dissociation from the doubly liganded dimer was rapid, but the second dissociation from the singly liganded dimer was slow. The dissociation rate constant from the liganded monomer was intermediate. The dynamic changes in the association and dissociation kinetics in relation to the dimerization of ErbB displayed negative cooperativity, which resulted in apparent low- and high-affinity sites of HRG association on the cell surface.


Experimental Dermatology | 2012

An ErbB receptor-mediated AP-1 regulatory network is modulated by STAT3 and c-MYC during calcium-dependent keratinocyte differentiation.

Yuko Saeki; Takeshi Nagashima; Shuhei Kimura; Mariko Okada-Hatakeyama

Abstract:  A calcium gradient in skin epidermis is known to regulate epidermal differentiation. In cultures of human epidermal keratinocytes (NHEK), induction of calcium‐dependent differentiation is associated with phosphorylation of ErbB receptors, including the epidermal growth factor receptor (EGFR). The activation of EGFR triggers the induction of activator protein 1 (AP‐1) proteins necessary for keratinocyte terminal differentiation. Interestingly, an in vitro long‐term calcium treatment revealed the activation of different ErbB receptors with different timings, which is consistent with the differential localization of each receptor in the skin layers in vivo. In the current study, the regulatory relationship between ErbB receptor activation and induction of AP‐1 proteins in calcium‐dependent keratinocyte differentiation was analysed. For this purpose, we used a mathematical method to predict molecular regulations from time‐course proteomic data of 30 target components. The predicted network showed that the ErbB receptor might regulate AP‐1 protein expression via two pathways: positive regulation by c‐MYC and negative regulation by signal transducer and activator of transcription 3 (STAT3) pathways. Experimental validation analysis revealed that ErbB receptor inhibition resulted in defective induction of AP‐1 proteins and suppressed terminal differentiation of keratinocytes. Inhibition of STAT3, however, affected expression of a partial set of AP‐1 proteins and resulted in premature differentiation. Studies using RNAi to diminish expression of each ErbB receptor and the AP‐1 proteins strongly suggested that STAT3 established a balance that maintains the appropriate set of AP‐1 proteins and participates in a complex network for regulating keratinocyte differentiation.


BMC Systems Biology | 2011

Epidermal growth factor receptor mutation in combination with expression of MIG6 alters gefitinib sensitivity

Yoshimi Naruo; Takeshi Nagashima; Ryoko Ushikoshi-Nakayama; Yuko Saeki; Takashi Nakakuki; Takashi Naka; Hiroshi Tanaka; Shih-Feng Tsai; Mariko Okada-Hatakeyama

BackgroundEpidermal growth factor receptor (EGFR) signaling plays an important role in the regulation of cell proliferation, survival, metastasis, and invasion in various tumors. Earlier studies showed that the EGFR is frequently overexpressed in non-small-cell lung cancer (NSCLC) and EGFR mutations at specific amino acid residues in the kinase domain induce altered responsiveness to gefitinib, a small molecule EGFR tyrosine kinase inhibitor. However, the mechanism underlying the drug response modulated by EGFR mutation is still largely unknown. To elucidate drug response in EGFR signal transduction pathway in which complex dynamics of multiple molecules involved, a systematic approach is necessary. In this paper, we performed experimental and computational analyses to clarify the underlying mechanism of EGFR signaling and cell-specific gefitinib responsiveness in three H1299-derived NSCLC cell lines; H1299 wild type (H1299WT), H1299 with an overexpressed wild type EGFR (H1299EGFR-WT), and H1299 with an overexpressed mutant EGFR L858R (H1299L858R; gefitinib sensitive mutant).ResultsWe predicted and experimentally verified that Mig6, which is a known negative regulator of EGFR and specifically expressed in H1299L858R cells, synergized with gefitinib to suppress cellular growth. Computational analyses indicated that this inhibitory effect is amplified at the phosphorylation/dephosphorylation steps of MEK and ERK.ConclusionsThus, we showed that L858R receptor mutation in combination with expression of its negative regulator, Mig6, alters signaling outcomes and results in variable drug sensitivity.


FEBS Journal | 2015

Feedforward regulation of mRNA stability by prolonged extracellular signal‐regulated kinase activity

Takeshi Nagashima; Norihiko Inoue; Noriko Yumoto; Yuko Saeki; Shigeyuki Magi; Natalia Volinsky; Alexander Sorkin; Boris N. Kholodenko; Mariko Okada-Hatakeyama

Extracellular signal‐regulated kinase (ERK) plays a central role in signal transduction networks and cell fate decisions. Sustained ERK activation induces cell differentiation, whereas transient ERK results in the proliferation of several types of cells. Sustained ERK activity stabilizes the proteins of early‐response gene products. However, the effect of ERK activity duration on mRNA stability is unknown. We analyzed the quantitative relationship between the duration of four ERK activity kinetics and the mRNA expression profile in growth factor‐treated cells. Time‐course transcriptome analysis revealed that the cells with prolonged ERK activity generally showed sustained mRNA expression of late response genes but not early or mid genes. Selected late response genes decayed more rapidly in the presence of a specific ERK inhibitor than a general transcription inhibitor and the decay rate was not related to the number of AU‐rich elements. Our results suggest that sustained ERK activity plays an important role in the lifespan of the mRNA encoded by late response genes, in addition to the previously demonstrated role in protein stabilization of early‐response genes, including transcription factors regulating the transcription of mid and late genes. This double‐positive regulation of ligand‐induced genes, also termed feedforward regulation, is critical in cell fate decisions.


生物物理 | 2013

3P179 生細胞内における厳密なPI3Kヘテロダイマー複合体のシグナル応答(12.細胞生物的課題,ポスター,日本生物物理学会年会第51回(2013年度))

Chan-Gi Pack; Yuko Saeki; Mariko Okada; Yasushi Sako


Seibutsu Butsuri | 2013

3P179 Dynamic Signal Response of Rigorous PI3K Heterodimer in Living Cells(12. Cell biology,Poster)

Chan-Gi Pack; Yuko Saeki; Mariko Okada; Yasushi Sako


Journal of Dermatological Science | 2013

An ErbB receptor mediated AP-1 regulatory network is modulated by STAT3 and c-MYC during calcium dependent keratinocyte differentiation

Yuko Saeki; Takeshi Nagashima; Shuhei Kimura; Mariko Okada-Hatakeyama

Collaboration


Dive into the Yuko Saeki'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

Mariko Hatakeyama

National Institute of Advanced Industrial Science and Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hiroshi Tanaka

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yoshimi Naruo

Tokyo Medical and Dental University

View shared research outputs
Researchain Logo
Decentralizing Knowledge