Network


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

Hotspot


Dive into the research topics where Yasutaka Hayashi is active.

Publication


Featured researches published by Yasutaka Hayashi.


Leukemia | 2015

SETBP1 Mutations Drive Leukemic Transformation in ASXL1-Mutated MDS

Daichi Inoue; Jiro Kitaura; Hirotaka Matsui; Hsin-An Hou; Wen-Chien Chou; Akiko Nagamachi; Kimihito C. Kawabata; Katsuhiro Togami; Reina Nagase; Sayuri Horikawa; Makoto Saika; Jean-Baptiste Micol; Yasutaka Hayashi; Yuka Harada; Hironori Harada; Toshiya Inaba; Hwei-Fang Tien; Omar Abdel-Wahab; Toshio Kitamura

Mutations in ASXL1 are frequent in patients with myelodysplastic syndrome (MDS) and are associated with adverse survival, yet the molecular pathogenesis of ASXL1 mutations (ASXL1-MT) is not fully understood. Recently, it has been found that deletion of Asxl1 or expression of C-terminal-truncating ASXL1-MTs inhibit myeloid differentiation and induce MDS-like disease in mice. Here, we find that SET-binding protein 1 (SETBP1) mutations (SETBP1-MT) are enriched among ASXL1-mutated MDS patients and associated with increased incidence of leukemic transformation, as well as shorter survival, suggesting that SETBP1-MT play a critical role in leukemic transformation of MDS. We identify that SETBP1-MT inhibit ubiquitination and subsequent degradation of SETBP1, resulting in increased expression. Expression of SETBP1-MT, in turn, inhibited protein phosphatase 2A activity, leading to Akt activation and enhanced expression of posterior Hoxa genes in ASXL1-mutant cells. Biologically, SETBP1-MT augmented ASXL1-MT-induced differentiation block, inhibited apoptosis and enhanced myeloid colony output. SETBP1-MT collaborated with ASXL1-MT in inducing acute myeloid leukemia in vivo. The combination of ASXL1-MT and SETBP1-MT activated a stem cell signature and repressed the tumor growth factor-β signaling pathway, in contrast to the ASXL1-MT-induced MDS model. These data reveal that SETBP1-MT are critical drivers of ASXL1-mutated MDS and identify several deregulated pathways as potential therapeutic targets in high-risk MDS.


Leukemia | 2005

Novel missense mutations in the tyrosine kinase domain of the platelet-derived growth factor receptor |[alpha]|(PDGFRA) gene in childhood acute myeloid leukemia with t(8;21)(q22;q22) or inv(16)(p13q22)

Mitsuteru Hiwatari; Tomohiko Taki; Masahiro Tsuchida; Ryouji Hanada; Teruaki Hongo; Masahiro Sako; Yasutaka Hayashi

Novel missense mutations in the tyrosine kinase domain of the platelet-derived growth factor receptor α ( PDGFRA ) gene in childhood acute myeloid leukemia with t(8;21)(q22;q22) or inv(16)(p13q22)


Proceedings of the Japan Academy. Series B, Physical and Biological Sciences | 2014

The molecular basis of myeloid malignancies

Toshio Kitamura; Daichi Inoue; Naoko Okochi-Watanabe; Naoko Kato; Yukiko Komeno; Yang Lu; Yutaka Enomoto; Noriko Doki; Tomoyuki Uchida; Yuki Kagiyama; Katsuhiro Togami; Kimihito C. Kawabata; Reina Nagase; Sayuri Horikawa; Yasutaka Hayashi; Makoto Saika; Tomofusa Fukuyama; Kumi Izawa; Toshihiko Oki; Fumio Nakahara; Jiro Kitaura

Myeloid malignancies consist of acute myeloid leukemia (AML), myelodysplastic syndromes (MDS) and myeloproliferative neoplasm (MPN). The latter two diseases have preleukemic features and frequently evolve to AML. As with solid tumors, multiple mutations are required for leukemogenesis. A decade ago, these gene alterations were subdivided into two categories: class I mutations stimulating cell growth or inhibiting apoptosis; and class II mutations that hamper differentiation of hematopoietic cells. In mouse models, class I mutations such as the Bcr-Abl fusion kinase induce MPN by themselves and some class II mutations such as Runx1 mutations induce MDS. Combinations of class I and class II mutations induce AML in a variety of mouse models. Thus, it was postulated that hematopoietic cells whose differentiation is blocked by class II mutations would autonomously proliferate with class I mutations leading to the development of leukemia. Recent progress in high-speed sequencing has enabled efficient identification of novel mutations in a variety of molecules including epigenetic factors, splicing factors, signaling molecules and proteins in the cohesin complex; most of these are not categorized as either class I or class II mutations. The functional consequences of these mutations are now being extensively investigated. In this article, we will review the molecular basis of hematological malignancies, focusing on mouse models and the interfaces between these models and clinical findings, and revisit the classical class I/II hypothesis.


Leukemia | 1997

Alterations of p16 and p15 genes in acute leukemia with MLL gene rearrangements and their correlation with clinical features

Hiroaki Ohnishi; Shu-Xia Guo; Kohmei Ida; Tomohiko Taki; S Naritaka; Fumio Bessho; Masayoshi Yanagisawa; Ryoji Hanada; M Eguchi; N Kamada; Kenkichi Kita; Shunji Yamamori; Yasutaka Hayashi

p16 and p15 genes are putative tumor suppressor genes located on chromosome 9p21. In acute leukemias, alterations of p16 and p15 genes have been reported to occur exclusively in lymphoid lineage. We analyzed alterations of p16 and p15 genes in 46 acute leukemias with MLL gene rearrangements by Southern blot analysis, and investigated the association with clinical characteristics. We identified homozygous deletion of p16 and p15 genes in five (19%) of 27 acute lymphoblastic leukemias (ALLs) and in two (11%) of 19 acute myeloid leukemias (AMLs). Patients with homozygous deletion of p16 and p15 genes showed higher average leukocyte counts (343 × 109/l vs 271 × 109/l) and lower estimated 2-year survival rates than those with normal p16 and p15 genes (14.3 vs 30.7%), although the differences were not statistically significant. In addition, we investigated mutation of p16 gene by polymerase chain reaction single strand conformation polymorphism (PCR-SSCP) in 31 patients, but no mutation was found in the patients tested. Our results suggest that alterations of p16 and p15 genes are involved in a subset of acute leukemias with MLL gene rearrangement not only of lymphoid but also of myeloid phenotype. Homozygous deletion of p16 and p15 genes may be a possible adverse prognostic factor, although further analysis would be needed to confirm it.


Annals of Oncology | 2016

Spectrum of clinical and genetic features of patients with inherited platelet disorder with suspected predisposition to hematological malignancies: a nationwide survey in Japan

Akihide Yoshimi; Takashi Toya; Yasuhito Nannya; K. Takaoka; Keita Kirito; Etsuro Ito; Hideaki Nakajima; Yasutaka Hayashi; T. Takahashi; A. Moriya-Saito; Kenshi Suzuki; Hironori Harada; Norio Komatsu; Kensuke Usuki; Motoshi Ichikawa; Mineo Kurokawa

BACKGROUND Inherited thrombocytopenia (IT) contains several forms of familial thrombocytopenia and some of them have propensity to hematological malignancies. The etiological and genetic features of this heterogeneous syndrome have not yet been elucidated. PATIENTS AND METHODS We conducted a nationwide survey to collect clinical information and samples from patients with familial thrombocytopenia and/or hematological malignancies in order to obtain a comprehensive understanding of IT. RESULTS Among the 43 pedigrees with clinical samples, RUNX1 mutations were identified in 8 pedigrees (18.6%). While MYH9 and ANKRD26 mutations were identified in 2 and 1 pedigrees, respectively, no gene mutations were detected in the remaining 32 pedigrees from a panel of previously reported pathogenetic mutations. Clinical data were comparable between FPD/AML and non-FPD/AML probands. CONCLUSIONS Our study clarified that it is unexpectedly difficult to diagnose FPD/AML based on clinical information alone, and thus, genetic testing is strongly recommended. Our survey also identified some pedigrees with a strong family history of myelodysplastic syndromes of unknown origin. Additionally, there were 14 pedigrees in which three or more members were affected by immune thrombocytopenia (ITP), and a computer-aided simulation suggested that such a distribution almost never happens by coincidence, which implicates a genetic predisposition to ITP.


Journal of Experimental Medicine | 2018

Expression of mutant Asxl1 perturbs hematopoiesis and promotes susceptibility to leukemic transformation.

Reina Nagase; Daichi Inoue; Alessandro Pastore; Takeshi Fujino; Hsin-An Hou; Norimasa Yamasaki; Susumu Goyama; Makoto Saika; Akinori Kanai; Yasuyuki Sera; Sayuri Horikawa; Yasunori Ota; Shuhei Asada; Yasutaka Hayashi; Kimihito C. Kawabata; Reina Takeda; Hwei-Fang Tien; Hiroaki Honda; Omar Abdel-Wahab; Toshio Kitamura

Additional sex combs like 1 (ASXL1) is frequently mutated in myeloid malignancies and clonal hematopoiesis of indeterminate potential (CHIP). Although loss of ASXL1 promotes hematopoietic transformation, there is growing evidence that ASXL1 mutations might confer an alteration of function. In this study, we identify that physiological expression of a C-terminal truncated Asxl1 mutant in vivo using conditional knock-in (KI) results in myeloid skewing, age-dependent anemia, thrombocytosis, and morphological dysplasia. Although expression of mutant Asxl1 altered the functions of hematopoietic stem cells (HSCs), it maintained their survival in competitive transplantation assays and increased susceptibility to leukemic transformation by co-occurring RUNX1 mutation or viral insertional mutagenesis. KI mice displayed substantial reductions in H3K4me3 and H2AK119Ub without significant reductions in H3K27me3, distinct from the effects of Asxl1 loss. Chromatin immunoprecipitation followed by next-generation sequencing analysis demonstrated opposing effects of wild-type and mutant Asxl1 on H3K4me3. These findings reveal that ASXL1 mutations confer HSCs with an altered epigenome and increase susceptibility for leukemic transformation, presenting a novel model for CHIP.


Journal of Biochemistry | 2016

Novel working hypothesis for pathogenesis of hematological malignancies: combination of mutations-induced cellular phenotypes determines the disease (cMIP-DD).

Toshio Kitamura; Naoko Watanabe-Okochi; Yutaka Enomoto; Fumio Nakahara; Toshihiko Oki; Yukiko Komeno; Naoko Kato; Noriko Doki; Tomoyuki Uchida; Yuki Kagiyama; Katsuhiro Togami; Kimihito C. Kawabata; Koutarou Nishimura; Yasutaka Hayashi; Reina Nagase; Makoto Saika; Tsuyoshi Fukushima; Shuhei Asada; Takeshi Fujino; Yuto Izawa; Sayuri Horikawa; Tomofusa Fukuyama; Yosuke Tanaka; Ryoichi Ono; Susumu Goyama; Tetsuya Nosaka; Jiro Kitaura; Daichi Inoue

Recent progress in high-speed sequencing technology has revealed that tumors harbor novel mutations in a variety of genes including those for molecules involved in epigenetics and splicing, some of which were not categorized to previously thought malignancy-related genes. However, despite thorough identification of mutations in solid tumors and hematological malignancies, how these mutations induce cell transformation still remains elusive. In addition, each tumor usually contains multiple mutations or sometimes consists of multiple clones, which makes functional analysis difficult. Fifteen years ago, it was proposed that combination of two types of mutations induce acute leukemia; Class I mutations induce cell growth or inhibit apoptosis while class II mutations block differentiation, co-operating in inducing acute leukemia. This notion has been proven using a variety of mouse models, however most of recently found mutations are not typical class I/II mutations. Although some novel mutations have been found to functionally work as class I or II mutation in leukemogenesis, the classical class I/II theory seems to be too simple to explain the whole story. We here overview the molecular basis of hematological malignancies based on clinical and experimental results, and propose a new working hypothesis for leukemogenesis.


Nature Communications | 2018

Mutant ASXL1 cooperates with BAP1 to promote myeloid leukaemogenesis

Shuhei Asada; Susumu Goyama; Daichi Inoue; Shiori Shikata; Reina Takeda; Tsuyoshi Fukushima; Taishi Yonezawa; Takeshi Fujino; Yasutaka Hayashi; Kimihito Cojin Kawabata; Tomofusa Fukuyama; Yosuke Tanaka; Akihiko Yokoyama; Satoshi Yamazaki; Hiroko Kozuka-Hata; Masaaki Oyama; Shinya Kojima; Masahito Kawazu; Hiroyuki Mano; Toshio Kitamura

ASXL1 mutations occur frequently in myeloid neoplasms and are associated with poor prognosis. However, the mechanisms by which mutant ASXL1 induces leukaemogenesis remain unclear. In this study, we report mutually reinforcing effects between a C-terminally truncated form of mutant ASXL1 (ASXL1-MT) and BAP1 in promoting myeloid leukaemogenesis. BAP1 expression results in increased monoubiquitination of ASXL1-MT, which in turn increases the catalytic function of BAP1. This hyperactive ASXL1-MT/BAP1 complex promotes aberrant myeloid differentiation of haematopoietic progenitor cells and accelerates RUNX1-ETO-driven leukaemogenesis. Mechanistically, this complex induces upregulation of posterior HOXA genes and IRF8 through removal of H2AK119 ubiquitination. Importantly, BAP1 depletion inhibits posterior HOXA gene expression and leukaemogenicity of ASXL1-MT-expressing myeloid leukemia cells. Furthermore, BAP1 is also required for the growth of MLL-fusion leukemia cells with posterior HOXA gene dysregulation. These data indicate that BAP1, which has long been considered a tumor suppressor, in fact plays tumor-promoting roles in myeloid neoplasms.ASXL1 gene is often mutated in myeloid malignancies. Here, the authors show that mutant ASXL1 and BAP1 are in a positive feedback loop such that BAP1 induces monoubiquitination of mutant ASXL1, which in turn enhances BAP1 activity to potentiate myeloid transformation via HOXA clusters and IRF8.


Leukemia | 2018

High expression of ABCG2 induced by EZH2 disruption has pivotal roles in MDS pathogenesis

Kimihito C. Kawabata; Yasutaka Hayashi; Daichi Inoue; H Meguro; H Sakurai; T Fukuyama; Y Tanaka; S Asada; T Fukushima; Reina Nagase; R Takeda; Yuka Harada; Jiro Kitaura; Susumu Goyama; Hironori Harada; Hiroyuki Aburatani; Toshio Kitamura

Both proto-oncogenic and tumor-suppressive functions have been reported for enhancer of zeste homolog 2 (EZH2). To investigate the effects of its inactivation, a mutant EZH2 lacking its catalytic domain was prepared (EZH2-dSET). In a mouse bone marrow transplant model, EZH2-dSET expression in bone marrow cells induced a myelodysplastic syndrome (MDS)-like disease in transplanted mice. Analysis of these mice identified Abcg2 as a direct target of EZH2. Intriguingly, Abcg2 expression alone induced the same disease in the transplanted mice, where stemness genes were enriched. Interestingly, ABCG2 expression is specifically high in MDS patients. The present results indicate that ABCG2 de-repression induced by EZH2 mutations have crucial roles in MDS pathogenesis.


Blood | 1988

Cytogenetic findings and clinical features in acute leukemia and transient myeloproliferative disorder in Down's syndrome

Yasutaka Hayashi; Mariko Eguchi; Kenichi Sugita; Shinpei Nakazawa; Takeyuki Sato; Seiji Kojima; Fumio Bessho; S Konishi; Toshiya Inaba; Ryoji Hanada

Collaboration


Dive into the Yasutaka Hayashi'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
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge