Network


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

Hotspot


Dive into the research topics where Kimihito C. Kawabata is active.

Publication


Featured researches published by Kimihito C. Kawabata.


Journal of Clinical Investigation | 2013

Myelodysplastic syndromes are induced by histone methylation–altering ASXL1 mutations

Daichi Inoue; Jiro Kitaura; Katsuhiro Togami; Koutarou Nishimura; Yutaka Enomoto; Tomoyuki Uchida; Yuki Kagiyama; Kimihito C. Kawabata; Fumio Nakahara; Kumi Izawa; Toshihiko Oki; Akie Maehara; Masamichi Isobe; Akiho Tsuchiya; Yuka Harada; Hironori Harada; Takahiro Ochiya; Hiroyuki Aburatani; Hiroshi Kimura; Felicitas Thol; Michael Heuser; Ross L. Levine; Omar Abdel-Wahab; Toshio Kitamura

Recurrent mutations in the gene encoding additional sex combs-like 1 (ASXL1) are found in various hematologic malignancies and associated with poor prognosis. In particular, ASXL1 mutations are common in patients with hematologic malignancies associated with myelodysplasia, including myelodysplastic syndromes (MDSs), and chronic myelomonocytic leukemia. Although loss-of-function ASXL1 mutations promote myeloid transformation, a large subset of ASXL1 mutations is thought to result in stable truncation of ASXL1. Here we demonstrate that C-terminal–truncating Asxl1 mutations (ASXL1-MTs) inhibited myeloid differentiation and induced MDS-like disease in mice. ASXL1-MT mice displayed features of human-associated MDS, including multi-lineage myelodysplasia, pancytopenia, and occasional progression to overt leukemia. ASXL1-MT resulted in derepression of homeobox A9 (Hoxa9) and microRNA-125a (miR-125a) expression through inhibition of polycomb repressive complex 2–mediated (PRC2-mediated) methylation of histone H3K27. miR-125a reduced expression of C-type lectin domain family 5, member a (Clec5a), which is involved in myeloid differentiation. In addition, HOXA9 expression was high in MDS patients with ASXL1-MT, while CLEC5A expression was generally low. Thus, ASXL1-MT–induced MDS-like disease in mice is associated with derepression of Hoxa9 and miR-125a and with Clec5a dysregulation. Our data provide evidence for an axis of MDS pathogenesis that implicates both ASXL1 mutations and miR-125a as therapeutic targets in MDS.


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.


Blood | 2014

Hes1 promotes blast crisis in chronic myelogenous leukemia through MMP-9 upregulation in leukemic cells

Fumio Nakahara; Jiro Kitaura; Tomoyuki Uchida; Chiemi Nishida; Katsuhiro Togami; Daichi Inoue; Toshihiro Matsukawa; Yuki Kagiyama; Yutaka Enomoto; Kimihito C. Kawabata; Lai Chen-Yi; Yukiko Komeno; Kumi Izawa; Toshihiko Oki; Genta Nagae; Yuka Harada; Hironori Harada; Makoto Otsu; Hiroyuki Aburatani; Beate Heissig; Koichi Hattori; Toshio Kitamura

High levels of HES1 expression are frequently found in BCR-ABL(+) chronic myelogenous leukemia in blast crisis (CML-BC). In mouse bone marrow transplantation (BMT) models, co-expression of BCR-ABL and Hes1 induces CML-BC-like disease; however, the underlying mechanism remained elusive. Here, based on gene expression analysis, we show that MMP-9 is upregulated by Hes1 in common myeloid progenitors (CMPs). Analysis of promoter activity demonstrated that Hes1 upregulated MMP-9 by activating NF-κB. Analysis of 20 samples from CML-BC patients showed that MMP-9 was highly expressed in three, with two exhibiting high levels of HES1 expression. Interestingly, MMP-9 deficiency impaired the cobblestone area-forming ability of CMPs expressing BCR-ABL and Hes1 that were in conjunction with a stromal cell layer. In addition, CMPs expressing BCR-ABL and Hes1 secreted MMP-9, promoting the release of soluble Kit-ligand (sKitL) from stromal cells, thereby enhancing proliferation of the leukemic cells. In accordance, mice transplanted with CMPs expressing BCR-ABL and Hes1 exhibited high levels of sKitL as well as MMP-9 in the serum. Importantly, MMP-9 deficiency impaired the development of CML-BC-like disease induced by BCR-ABL and Hes1 in mouse BMT models. The present results suggest that Hes1 promotes the development of CML-BC, partly through MMP-9 upregulation in leukemic cells.


Experimental Hematology | 2015

A C-terminal mutant of CCAAT-enhancer-binding protein α (C/EBPα-Cm) downregulates Csf1r, a potent accelerator in the progression of acute myeloid leukemia with C/EBPα-Cm.

Katsuhiro Togami; Jiro Kitaura; Tomoyuki Uchida; Daichi Inoue; Koutarou Nishimura; Kimihito C. Kawabata; Reina Nagase; Sayuri Horikawa; Kumi Izawa; Tomofusa Fukuyama; Fumio Nakahara; Toshihiko Oki; Yuka Harada; Hironori Harada; Hiroyuki Aburatani; Toshio Kitamura

Two types of CCAAT-enhancer-binding protein α (C/EBPα) mutants are found in acute myeloid leukemia (AML) patients: N-terminal frame-shift mutants (C/EBPα-N(m)) generating p30 as a dominant form and C-terminal basic leucine zipper domain mutants (C/EBPα-C(m)). We have previously shown that C/EBPα-K304_R323dup belonging to C/EBPα-C(m), but not C/EBPα-T60fsX159 belonging to C/EBPα-N(m), alone induced AML in mouse bone marrow transplantation (BMT) models. Here we show that various C/EBPα-C(m) mutations have a similar, but not identical, potential in myeloid leukemogenesis. Notably, like C/EBPα-K304_R323dup, any type of C/EBPα-C(m) tested (C/EBPα-S299_K304dup, K313dup, or N321D) by itself induced AML, albeit with different latencies after BMT; C/EBPα-N321D induced AML with the shortest latency. By analyzing the gene expression profiles of C/EBPα-N321D- and mock-transduced c-kit(+)Sca-1(+)Lin(-) cells, we identified Csf1r as a gene downregulated by C/EBPα-N321D. In addition, leukemic cells expressing C/EBPα-C(m) exhibited low levels of colony stimulating factor 1 receptor in mice. On the other hand, transduction with C/EBPα-N(m) did not influence Csf1r expression in c-kit(+)Sca-1(+)Lin(-) cells, implying a unique role for C/EBPα-C(m) in downregulating Csf1r. Importantly, Csf1r overexpression collaborated with C/EBPα-N321D to induce fulminant AML with leukocytosis in mouse BMT models to a greater extent than did C/EBPα-N321D alone. Collectively, these results suggest that C/EBPα-C(m)-mediated downregulation of Csf1r has a negative, rather than a positive, impact on the progression of AML involving C/EBPα-C(m), which might possibly be accelerated by additional genetic and/or epigenetic alterations inducing Csf1r upregulation.


International Journal of Hematology | 2012

Upregulation of CD200R1 in lineage-negative leukemic cells is characteristic of AML1-ETO-positive leukemia in mice

Yuki Kagiyama; Jiro Kitaura; Katsuhiro Togami; Tomoyuki Uchida; Daichi Inoue; Toshihiro Matsukawa; Kumi Izawa; Kimihito C. Kawabata; Yukiko Komeno; Toshihiko Oki; Fumio Nakahara; Katsuaki Sato; Hiroyuki Aburatani; Toshio Kitamura

Activating mutations of c-Kit are frequently found in acute myeloid leukemia (AML) patients harboring t(8;21) chromosomal translocation generating a fusion protein AML1-ETO. Here we show that an active mutant of c-Kit cooperates with AML1-ETO to induce AML in mouse bone marrow transplantation models. Leukemic cells expressing AML1-ETO with c-KitD814V were serially transplantable. Transplantation experiments indicated that lineage−c-Kit+Sca-1+ (KSL) leukemic cells, but not lineage+ leukemic cells, were enriched for leukemia stem cells (LSCs). Comparison of gene expression profiles between KSL leukemic and normal cells delineated that CD200R1 was highly expressed in KSL leukemic cells as compared with KSL normal cells. Upregulation of CD200R1 was verified in lineage− leukemic cells, but not in lineage+ leukemic cells. CD200R1 expression in the lineage− leukemic cells was not correlated with the frequency of LSCs, indicating that CD200R1 is not a useful marker for LSCs in these models. Interestingly, CD200R1 was upregulated in KSL cells transduced with AML1-ETO, but not with other leukemogenic mutants, including c-KitD814V, AML1D171N, and AML1S291fsX300. Consistently, upregulation of CD200R1 in lineage− leukemic cells was observed only in the BM of mice suffering from AML1-ETO-positive leukemia. In conclusion, AML1-ETO upregulated CD200R1 in lineage− cells, which was characteristic of AML1-ETO-positive leukemia in mice.


Pulmonary circulation | 2015

Role played by Prx1-dependent extracellular matrix properties in vascular smooth muscle development in embryonic lungs

Kaori Ihida-Stansbury; Juliana Ames; Mithil Chokshi; Norman Aiad; Sonali Sanyal; Kimihito C. Kawabata; Ilya Levental; Harini G. Sundararaghavan; Jason A. Burdick; Paul A. Janmey; Kohei Miyazono; Rebecca G. Wells; Peter Lloyd Jones

Although there are many studies focusing on the molecular pathways underlying lung vascular morphogenesis, the extracellular matrix (ECM)–dependent regulation of mesenchymal cell differentiation in vascular smooth muscle development needs better understanding. In this study, we demonstrate that the paired related homeobox gene transcription factor Prx1 maintains the elastic ECM properties, which are essential for vascular smooth muscle precursor cell differentiation. We have found that Prx1null mouse lungs exhibit defective vascular smooth muscle development, downregulated elastic ECM expression, and compromised transforming growth factor (TGF)–β localization and signaling. Further characterization of ECM properties using decellularized lung ECM scaffolds derived from Prx1 mice demonstrated that Prx1 is required to maintain lung ECM stiffness. The results of cell culture using stiffness-controlled 2-D and 3-D synthetic substrates confirmed that Prx1-dependent ECM stiffness is essential for promotion of smooth muscle precursor differentiation for effective TGF-β stimulation. Supporting these results, both decellularized Prx1null lung ECM and Prx1WT (wild type) ECM scaffolds with blocked TGF-β failed to support mesenchymal cell to 3-D smooth muscle cell differentiation. These results suggest a novel ECM-dependent regulatory pathway of lung vascular development wherein Prx1 regulates lung vascular smooth muscle precursor development by coordinating the ECM biophysical and biochemical properties.


Experimental Hematology | 2014

Hes1 upregulation contributes to the development of FIP1L1-PDGRA-positive leukemia in blast crisis.

Tomoyuki Uchida; Jiro Kitaura; Fumio Nakahara; Katsuhiro Togami; Daichi Inoue; Akie Maehara; Koutarou Nishimura; Kimihito C. Kawabata; Noriko Doki; Kazuhiko Kakihana; Kosuke Yoshioka; Kumi Izawa; Toshihiko Oki; Akiko Sada; Yuka Harada; Kazuteru Ohashi; Yoshio Katayama; Toshimitsu Matsui; Hironori Harada; Toshio Kitamura

We have previously shown that elevated expression of Hairy enhancer of split 1 (Hes1) contributes to blast crisis transition in Bcr-Abl-positive chronic myelogenous leukemia. Here we investigate whether Hes1 is involved in the development of other myeloid neoplasms. Notably, Hes1 expression was elevated in only a few cases of 65 samples with different types of myeloid neoplasms. Interestingly, elevated expression of Hes1 was found in two of five samples of Fip1-like1 platelet-derived growth factor receptor-α (FIP1L1-PDGFA)-positive myeloid neoplasms associated with eosinophilia. Whereas FIP1L1-PDGFRα alone induced acute T-cell leukemia or myeloproliferative neoplasms in mouse bone marrow transplantation models, mice transplanted with bone marrow cells expressing both Hes1 and FIP1L1-PDGFRα developed acute leukemia characterized by an expansion of myeloid blasts and leukemic cells without eosinophilic granules. FIP1L1-PDGFRα conferred cytokine-independent growth to Hes1-transduced common myeloid progenitors, interleukin-3-dependent cells. Imatinib inhibited the growth of common myeloid progenitors expressing Hes1 with FIP1L1-PDGFRα, but not with imatinib-resistant FIP1L1-PDGFRα mutants harboring T674I or D842V. In contrast, ponatinib efficiently eradicated leukemic cells expressing Hes1 and the imatinib-resistant FLP1L1-PDGFRΑ mutant in vitro and in vivo. Thus, we have established mouse models of FIP1L1-PDGFRA-positive leukemia in myeloid blast crisis, which will help elucidate the pathogenesis of the disease and develop a new treatment for it.


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.

Collaboration


Dive into the Kimihito C. Kawabata'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