Network


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

Hotspot


Dive into the research topics where Makoto Saika is active.

Publication


Featured researches published by Makoto Saika.


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.


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.


Experimental Hematology | 2016

Truncation mutants of ASXL1 observed in myeloid malignancies are expressed at detectable protein levels

Daichi Inoue; Masaki Matsumoto; Reina Nagase; Makoto Saika; Takeshi Fujino; Keiichi I. Nakayama; Toshio Kitamura

Recent progress in deep sequencing technologies has revealed many novel mutations in a variety of genes in patients with myelodysplastic syndromes (MDS). Most of these mutations are thought to be loss-of-function mutations, with some exceptions, such as the gain-of-function IDH1/2 and SRSF2 mutations. Among the mutations, ASXL1 mutations attract much attention; the ASXL1 mutations are identified in a variety of hematologic malignancies and always predicts poor prognosis. It was found that the C-terminal truncating mutants of the ASXL1 or ASXL1 deletion induced MDS-like diseases in mouse. In addition, it has recently been reported that ASXL1 mutations are frequently found in clonal hematopoiesis in healthy elderly people, who frequently progress to hematologic malignancies. However, the underlying molecular mechanisms by which ASXL1 mutations induce hematologic malignancies are not fully understood. Moreover, whether ASXL1 mutations are loss-of-function mutations or dominant-negative or gain-of-function mutations remains a matter of controversy. We here present solid evidence indicating that the C-terminal truncating ASXL1 protein is indeed expressed in cells harboring homozygous mutations of ASXL1, indicating the ASXL1 mutations are dominant-negative or gain-of-function mutations; for the first time, we detected the truncated ASXL1 proteins in two cell lines lacking the intact ASXL1 gene by mass spectrometry and Western blot analyses. Thus, together with our previous results, the present results indicate that the truncating ASXL1 mutant is indeed expressed in MDS cells and may play a role in MDS pathogenesis not previously considered.


Leukemia | 2018

A novel ASXL1–OGT axis plays roles in H3K4 methylation and tumor suppression in myeloid malignancies

Daichi Inoue; Takeshi Fujino; Paul Sheridan; Yao-zhong Zhang; Reina Nagase; Sayuri Horikawa; Zaomin Li; Hirotaka Matsui; Akinori Kanai; Makoto Saika; Rui Yamaguchi; Hiroko Kozuka-Hata; Kimihito C. Kawabata; Akihiko Yokoyama; Susumu Goyama; Toshiya Inaba; Seiya Imoto; Satoru Miyano; Mingjiang Xu; Feng Chun Yang; Masaaki Oyama; Toshio Kitamura

ASXL1 plays key roles in epigenetic regulation of gene expression through methylation of histone H3K27, and disruption of ASXL1 drives myeloid malignancies, at least in part, via derepression of posterior HOXA loci. However, little is known about the identity of proteins that interact with ASXL1 and about the functions of ASXL1 in modulation of the active histone mark, such as H3K4 methylation. In this study, we demonstrate that ASXL1 is a part of a protein complex containing HCFC1 and OGT; OGT directly stabilizes ASXL1 by O-GlcNAcylation. Disruption of this novel axis inhibited myeloid differentiation and H3K4 methylation as well as H2B glycosylation and impaired transcription of genes involved in myeloid differentiation, splicing, and ribosomal functions; this has implications for myelodysplastic syndrome (MDS) pathogenesis, as each of these processes are perturbed in the disease. This axis is responsible for tumor suppression in the myeloid compartment, as reactivation of OGT induced myeloid differentiation and reduced leukemogenecity both in vivo and in vitro. Our data also suggest that MLL5, a known HCFC1/OGT-interacting protein, is responsible for gene activation by the ASXL1–OGT axis. These data shed light on the novel roles of the ASXL1–OGT axis in H3K4 methylation and activation of transcription.


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.


Scientific Reports | 2018

ASXL1 and SETBP1 mutations promote leukaemogenesis by repressing TGFβ pathway genes through histone deacetylation

Makoto Saika; Daichi Inoue; Reina Nagase; Naru Sato; Akiho Tsuchiya; Tomohiro Yabushita; Toshio Kitamura; Susumu Goyama

Mutations in ASXL1 and SETBP1 genes have been frequently detected and often coexist in myelodysplastic syndrome (MDS) and acute myeloid leukaemia (AML). We previously showed that coexpression of mutant ASXL1 and SETBP1 in hematopoietic progenitor cells induced downregulation of TGFβ pathway genes and promoted the development of MDS/AML in a mouse model of bone marrow transplantation. However, whether the repression of TGFβ pathway in fact contributes to leukaemogenesis remains unclear. Moreover, mechanisms for the repression of TGFβ pathway genes in ASXL1/SETBP1-mutated MDS/AML cells have not been fully understood. In this study, we showed that expression of a constitutively active TGFβ type I receptor (ALK5-TD) inhibited leukaemic proliferation of MDS/AML cells expressing mutant ASXL1/SETBP1. We also found aberrantly reduced acetylation of several lysine residues on histone H3 and H4 around the promoter regions of multiple TGFβ pathway genes. The histone deacetylase (HDAC) inhibitor vorinostat reversed histone acetylation at these promoter regions, and induced transcriptional derepression of the TGFβ pathway genes. Furthermore, vorinostat showed robust growth-inhibitory effect in cells expressing mutant ASXL1, whereas it showed only a marginal effect in normal bone marrow cells. These data indicate that HDAC inhibitors will be promising therapeutic drugs for MDS and AML with ASXL1 and SETBP1 mutations.


Experimental Hematology | 2016

Analysis of MDS mice model induced by ASXL1 and RUNX1 mutants

Reina Nagase; Daichi Inoue; Makoto Saika; Hsin-An Hou; Wen-Chien Chou; Kimihito C. Kawabata; Hironori Harada; Akinori Kanai; Susumu Goyama; Hiroaki Honda; Hwei-Fang Tien; Toshio Kitamura


Experimental Hematology | 2015

Novel roles of ASXL1 in epigenetic regulation

Daichi Inoue; Sayuri Horikawa; Hirotaka Matsui; Reina Nagase; Makoto Saika; Kimihito C. Kawabata; Susumu Goyama; Toshio Kitamura


The Japanese journal of clinical hematology | 2014

Molecular basis of hematological malignancies

Toshio Kitamura; Daichi Inoue; Fumio Nakahara; Okochi N; Naoko Kato; Katsuhiro Togami; Toshiki Uchida; Yuki Kagiyama; Kimihito C. Kawabata; Reina Nagase; Sayuri Horikawa; Hayashi K; Makoto Saika; Kumi Izawa; Toshihiko Oki; Shigeru Chiba; Yuichi Harada; Hironori Harada; Jiro Kitaura

Collaboration


Dive into the Makoto Saika'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