Network


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

Hotspot


Dive into the research topics where Yoichi Imai is active.

Publication


Featured researches published by Yoichi Imai.


Molecular and Cellular Biology | 1996

The extracellular signal-regulated kinase pathway phosphorylates AML1, an acute myeloid leukemia gene product, and potentially regulates its transactivation ability.

Tomoyuki Tanaka; Mineo Kurokawa; Kohjiro Ueki; Kozo Tanaka; Yoichi Imai; Kinuko Mitani; Kenji Okazaki; Noriyuki Sagata; Yoshio Yazaki; Yoichi Shibata; Takashi Kadowaki; Hisamaru Hirai

AML1 (also called PEBP2alphaB, CBFA2, or CBFalpha2) is one of the most frequently disrupted genes in chromosome abnormalities seen in human leukemias. It has been reported that AML1 plays several pivotal roles in myeloid hematopoietic differentiation and other biological phenomena, probably through the transcriptional regulation of various relevant genes. Here, we investigated the mechanism of regulation of AML1 functions through signal transduction pathways. The results showed that AML1 is phosphorylated in vivo on two serine residues within the proline-, serine-, and threonine-rich region, with dependence on the activation of extracellular signal-regulated kinase (ERK) and with interleukin-3 stimulation in a hematopoietic cell line. These in vivo phosphorylation sites of AML1 were phosphorylated directly in vitro by ERK. Although differences between wild-type AML1 and phosphorylation site mutants in DNA-binding affinity were not observed, we have shown that ERK-dependent phosphorylation potentiates the transactivation ability of AML1. Furthermore the phosphorylation site mutations reduced the transforming capacity of AML1 in fibroblast cells. These data indicate that AML1 functions are potentially regulated by ERK, which is activated by cytokine and growth factor stimuli. This study provides some important clues for clarifying unidentified facets of the regulatory mechanism of AML1 function.


Biology of Blood and Marrow Transplantation | 2009

Influence of Pretransplantation Serum Ferritin on Nonrelapse Mortality after Myeloablative and Nonmyeloablative Allogeneic Hematopoietic Stem Cell Transplantation

Keisuke Kataoka; Yasuhito Nannya; Akira Hangaishi; Yoichi Imai; Shigeru Chiba; Tsuyoshi Takahashi; Mineo Kurokawa

Iron overload might be an important contributor to nonrelapse mortality (NRM) in hematopoietic stem cell transplantation (HSCT). We studied 264 patients undergoing allogeneic HSCT for hematologic malignancies between 1996 and 2006, using pretransplantation serum ferritin as a surrogate marker of iron overload. At 5 years, patients in the high ferritin group (>or= 599 ng/mL) had a lower overall survival (OS; 33.0% versus 63.5%; P< .001) and a higher NRM (34.9% versus 13.7%; P< .001) than those in the low ferritin group (<599 ng/mL). Multivariate analyses showed that high pretransplantation serum ferritin was a significant risk factor for worse survival (relative risk [RR]=1.68; P= .05) and increased NRM (RR=2.47; P= .01). There was no significant difference in the cumulative incidence of relapse, and acute and chronic graft-versus-host disease (aGVHD, cGVHD) between the 2 groups. Patients in the high ferritin group were more likely to die of infection (P< .010) and organ failure (P< .019). Similar results were observed after dividing the patients according to the intensity of conditioning regimens. These findings emphasize the prognostic impact of pretransplantation serum ferritin in HSCT recipients.


Blood | 2011

Evi1 represses PTEN expression and activates PI3K/AKT/mTOR via interactions with polycomb proteins

Akihide Yoshimi; Susumu Goyama; Naoko Watanabe-Okochi; Yumiko Yoshiki; Yasuhito Nannya; Eriko Nitta; Shunya Arai; Tomohiko Sato; Munetake Shimabe; Masahiro Nakagawa; Yoichi Imai; Toshio Kitamura; Mineo Kurokawa

Evi1 (ecotropic viral integration site 1) is essential for proliferation of hematopoietic stem cells and implicated in the development of myeloid disorders. Particularly, high Evi1 expression defines one of the largest clusters in acute myeloid leukemia and is significantly associated with extremely poor prognosis. However, mechanistic basis of Evi1-mediated leukemogenesis has not been fully elucidated. Here, we show that Evi1 directly represses phosphatase and tensin homologue deleted on chromosome 10 (PTEN) transcription in the murine bone marrow, which leads to activation of AKT/mammalian target of rapamycin (mTOR) signaling. In a murine bone marrow transplantation model, Evi1 leukemia showed modestly increased sensitivity to an mTOR inhibitor rapamycin. Furthermore, we found that Evi1 binds to several polycomb group proteins and recruits polycomb repressive complexes for PTEN down-regulation, which shows a novel epigenetic mechanism of AKT/mTOR activation in leukemia. Expression analyses and ChIPassays with human samples indicate that our findings in mice models are recapitulated in human leukemic cells. Dependence of Evi1-expressing leukemic cells on AKT/mTOR signaling provides the first example of targeted therapeutic modalities that suppress the leukemogenic activity of Evi1. The PTEN/AKT/mTOR signaling pathway and the Evi1-polycomb interaction can be promising therapeutic targets for leukemia with activated Evi1.


Journal of Experimental Medicine | 2011

Evi1 is essential for hematopoietic stem cell self-renewal, and its expression marks hematopoietic cells with long-term multilineage repopulating activity.

Keisuke Kataoka; Tomohiko Sato; Akihide Yoshimi; Susumu Goyama; Takako Tsuruta; Hiroshi Kobayashi; Munetake Shimabe; Shunya Arai; Masahiro Nakagawa; Yoichi Imai; Keiki Kumano; Katsuyoshi Kumagai; Naoto Kubota; Takashi Kadowaki; Mineo Kurokawa

A new mouse in which an IRES-GFP cassette is knocked-in to the Evi1 locus reveals that HSC long-term multilineage repopulating activity specifically segregates with expression of the Evi1 transcription factor.


Oncogene | 2001

Mutations of the Smad4 gene in acute myelogeneous leukemia and their functional implications in leukemogenesis

Yoichi Imai; Mineo Kurokawa; Koji Izutsu; Akira Hangaishi; Kazuhiro Maki; Seishi Ogawa; Shigeru Chiba; Kinuko Mitani; Hisamaru Hirai

The Smad family proteins are critical components of the transforming growth factor (TGF)-β signaling pathway. TGF-β is a multipotent cytokine that elicits many biological functions. In particular, TGF-β exhibits effects on the cell cycle manifested by G1-phase arrest, differentiation, or apoptosis of several target cells, suggesting that disruption of TGF-β signaling pathway could be involved in cancer formation. Here we show one missense mutation of the Smad4 gene in the MH1 domain (P102L) and one frame shift mutation resulting in termination in the MH2 domain (Δ(483–552)) in acute myelogeneous leukemia. Both of the mutated Smad4 proteins lack transcriptional activities. Concomitant expression of the P102L mutant with wild-type Smad4 inactivates wild-type Smad4 through inhibiting its DNA-binding ability. The Δ(483–552) mutant blocks nuclear translocation of wild-type Smad4 and thus disrupts TGF-β signaling. This is the first report showing that mutations in the Smad4 gene are associated with the pathogenesis of acute myelogeneous leukemia and the obtained results should provide useful insights into the mechanism whereby disruption of TGF-β signaling pathway could lead to acute myelogeneous leukemia.


Molecular and Cellular Biology | 2004

The Corepressor mSin3A Regulates Phosphorylation-Induced Activation, Intranuclear Location, and Stability of AML1

Yoichi Imai; Mineo Kurokawa; Yuko Yamaguchi; Koji Izutsu; Eriko Nitta; Kinuko Mitani; Masanobu Satake; Tetsuo Noda; Yoshiaki Ito; Hisamaru Hirai

ABSTRACT The AML1 (RUNX1) gene, one of the most frequent targets of translocations associated with human leukemias, encodes a DNA-binding protein that plays pivotal roles in myeloid differentiation through transcriptional regulation of various genes. Previously, we reported that AML1 is phosphorylated on two serine residues with dependence on activation of extracellular signal-regulated kinase, which positively regulates the transcriptional activity of AML1. Here, we demonstrate that the interaction between AML1 and the corepressor mSin3A is regulated by phosphorylation of AML1 and that release of AML1 from mSin3A induced by phosphorylation activates its transcriptional activity. Furthermore, phosphorylation of AML1 regulates its intranuclear location and disrupts colocalization of AML1 with mSin3A in the nuclear matrix. PEBP2β/CBFβ, a heterodimeric partner of AML1, was shown to play a role in protecting AML1 from proteasome-mediated degradation. We show that mSin3A also protects AML1 from proteasome-mediated degradation and that phosphorylation-induced release of AML1 from mSin3A results in degradation of AML1 in a time-dependent manner. This study provides a novel regulatory mechanism for the function of transcription factors mediated by protein modification and interaction with cofactors.


Blood | 2011

Evi-1 is a transcriptional target of mixed-lineage leukemia oncoproteins in hematopoietic stem cells

Shunya Arai; Akihide Yoshimi; Munetake Shimabe; Motoshi Ichikawa; Masahiro Nakagawa; Yoichi Imai; Susumu Goyama; Mineo Kurokawa

Ecotropic viral integration site-1 (Evi-1) is a nuclear transcription factor that plays an essential role in the regulation of hematopoietic stem cells. Aberrant expression of Evi-1 has been reported in up to 10% of patients with acute myeloid leukemia and is a diagnostic marker that predicts a poor outcome. Although chromosomal rearrangement involving the Evi-1 gene is one of the major causes of Evi-1 activation, overexpression of Evi-1 is detected in a subgroup of acute myeloid leukemia patients without any chromosomal abnormalities, which indicates the presence of other mechanisms for Evi-1 activation. In this study, we found that Evi-1 is frequently up-regulated in bone marrow cells transformed by the mixed-lineage leukemia (MLL) chimeric genes MLL-ENL or MLL-AF9. Analysis of the Evi-1 gene promoter region revealed that MLL-ENL activates transcription of Evi-1. MLL-ENL-mediated up-regulation of Evi-1 occurs exclusively in the undifferentiated hematopoietic population, in which Evi-1 particularly contributes to the propagation of MLL-ENL-immortalized cells. Furthermore, gene-expression analysis of human acute myeloid leukemia cases demonstrated the stem cell-like gene-expression signature of MLL-rearranged leukemia with high levels of Evi-1. Our findings indicate that Evi-1 is one of the targets of MLL oncoproteins and is selectively activated in hematopoietic stem cell-derived MLL leukemic cells.


Cancer Science | 2005

Notch1 oncoprotein antagonizes TGF‐β/Smad‐mediated cell growth suppression via sequestration of coactivator p300

Shigeo Masuda; Keiki Kumano; Kiyoshi Shimizu; Yoichi Imai; Mineo Kurokawa; Seishi Ogawa; Makoto Miyagishi; Kazunari Taira; Hisamaru Hirai; Shigeru Chiba

The Notch proteins constitute a family of transmembrane receptors that play a pivotal role in cellular differentiation, proliferation and apoptosis. Although it has been recognized that excess Notch signaling is potentially tumorigenic, little is known about precise mechanisms through which dysregulated Notch signaling induces neoplastic transformation. Here we demonstrate that Notch signaling has a transcriptional cross‐talk with transforming growth factor‐β (TGF‐β) signaling, which is well characterized by its antiproliferative effects. TGF‐β‐mediated transcriptional responses are suppressed by constitutively active Notch1, and this inhibitory effect is canceled by introduction of transcriptional coactivator p300. We further show that this blockade of TGF‐β signaling is executed by the sequestration of p300 from Smad3. Moreover, in a human cervical carcinoma cell line, CaSki, in which Notch1 is spontaneously activated, suppression of Notch1 expression with small interfering RNA significantly restores the responsiveness to TGF‐β. Taken together, we propose that Notch oncoproteins promote cell growth and cancer development partly by suppressing the growth inhibitory effects of TGF‐β through sequestrating p300 from Smad3. (Cancer Sci 2005; 96: 274 –283)


Oncogene | 2005

Oligomerization of Evi-1 regulated by the PR domain contributes to recruitment of corepressor CtBP

Eriko Nitta; Koji Izutsu; Yuko Yamaguchi; Yoichi Imai; Seishi Ogawa; Shigeru Chiba; Mineo Kurokawa; Hisamaru Hirai

Evi-1 is a transcription factor that is implicated in leukemic transformation of hematopoietic cells. Two distinct alternative forms, Evi-1a and Evi-1c, are generated from the EVI-1 gene. Whereas Evi-1a is widely recognized as an oncoprotein, a role for Evi-1c, which has an additional PR domain in the amino-terminus of Evi-1a, in leukemogenesis, has not been elucidated thus far. Aberrant oligomerization of transcription factors has recently emerged as a prevalent mechanism for activating their oncogenic potential in hematopoietic malignancies. Here, to study the mechanisms that underlie Evi-1-mediated oncogenesis, we investigated formation of oligomeric complexes by the Evi-1 proteins. We show that Evi-1a forms homo-oligomers, whereas Evi-1c exclusively exists as a monomer in mammalian cells. Remarkably, Evi-1c has lost the ability to interact with CtBP, a transcriptional corepressor that associates with Evi-1a. As a consequence, the ability of Evi-1c to repress transforming growth factor-β (TGF-β) signaling is significantly abrogated. These results identify a novel function of a PR domain to regulate oligomerization of transcription factors and suggest that homo-oligomerization may play a critical role in corepressor recruitment by the Evi-1 proteins. In addition, we found that the chimeric oncoprotein acute myelocytic leukemia (AML)1-Evi-1, generated in t(3;21) leukemia, also forms homo-oligomers and hetero-oligomers with Evi-1a, while it did not interact with Evi-1c. Consistent with the results, repression of TGF-β by AML1-Evi-1 was significantly enhanced by Evi-1a, whereas it was hardly affected by the presence of Evi-1c. These results suggest that oligomerization may contribute to the oncogenic potential of Evi-1-containing proteins.


Oncogene | 2009

Pbx1 is a downstream target of Evi-1 in hematopoietic stem/progenitors and leukemic cells.

Munetake Shimabe; Susumu Goyama; Naoko Watanabe-Okochi; Akihide Yoshimi; Motoshi Ichikawa; Yoichi Imai; Mineo Kurokawa

Ecotropic viral integration site-1 (Evi-1) is a nuclear transcription factor, which is essential for the proliferation/maintenance of hematopoietic stem cells (HSCs). Aberrant expression of Evi-1 has been frequently found in myeloid leukemia, and is associated with a poor patient survival. Recently, we reported candidate target genes of Evi-1 shared in HSCs and leukemic cells using gene expression profiling analysis. In this study, we identified Pbx1, a proto-oncogene in hematopoietic malignancy, as a target gene of Evi-1. Overexpression of Evi-1 increased Pbx1 expression in hematopoietic stem/progenitor cells. An analysis of the Pbx1 promoter region revealed that Evi-1 upregulates Pbx1 transcription. Furthermore, reduction of Pbx1 levels through RNAi-mediated knockdown significantly inhibited Evi-1-induced transformation. In contrast, knockdown of Pbx1 did not impair bone marrow transformation by E2A/HLF or AML1/ETO, suggesting that Pbx1 is specifically required for the maintenance of bone marrow transformation mediated by Evi-1. These results indicate that Pbx1 is a target gene of Evi-1 involved in Evi-1-mediated leukemogenesis.

Collaboration


Dive into the Yoichi Imai'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

Kinuko Mitani

Dokkyo Medical University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge