研究者業績

砂河 孝行

Isagawa Takayuki

基本情報

所属
自治医科大学 データサイエンスセンター 講師
(兼任)分子病態治療研究センター循環病態・代謝学研究部 講師
学位
博士(理学)

J-GLOBAL ID
201801008225774969
researchmap会員ID
B000316243

論文

 61
  • Nobuaki Shiraki, Yuichiro Higuchi, Seiko Harada, Kahoko Umeda, Takayuki Isagawa, Hiroyuki Aburatani, Kazuhiko Kume, Shoen Kume
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 381(4) 694-699 2009年4月  査読有り
    Embryonic stem cells differentiated on M15 cells have previously been shown to give rise to cells of the mesendodermal and definitive endodermal lineages. Here we demonstrate that neuroectodermal and mesodermal lineages can be derived from ES cells cultured on M15 cells and subsequently subjected to specific culture conditions, as confirmed by the expression of molecular markers. Prospective isolation and microarray analyses showed that neuroectodermal cells expressed anterior-to-posterior, as well as dorso-ventral regional markers, suggesting that this procedure could be used for the induction of cells belonging to a wide variety of neural lineages. Lateral mesoderm and paraxial mesoderm cells were also produced and their gene expression profiles were confirmed by microarray analyses. These results indicate that the M15 cell system provides a valuable tool for generating ES cell-derived lineage-specific cell types belonging to the three germ layers, namely neuroectoderm, mesoderm, and definitive endoderm. (C) 2009 Elsevier Inc. All rights reserved.
  • Kenji Matsumoto, Takayuki Isagawa, Toshinobu Nishimura, Takunori Ogaeri, Koji Eto, Satsuki Miyazaki, Jun-ichi Miyazaki, Hiroyuki Aburatani, Hiromitsu Nakauchi, Hideo Ema
    PLOS ONE 4(3) e4820 2009年3月  査読有り
    The cellular ontogeny of hematopoietic stem cells (HSCs) remains poorly understood because their isolation from and their identification in early developing small embryos are difficult. We attempted to dissect early developmental stages of HSCs using an in vitro mouse embryonic stem cell (ESC) differentiation system combined with inducible HOXB4 expression. Here we report the identification of pre-HSCs and an embryonic type of HSCs (embryonic HSCs) as intermediate cells between ESCs and HSCs. Both pre-HSCs and embryonic HSCs were isolated by their c-Kit+ CD41+ CD45 2 phenotype. Pre-HSCs did not engraft in irradiated adult mice. After co-culture with OP9 stromal cells and conditional expression of HOXB4, pre-HSCs gave rise to embryonic HSCs capable of engraftment and long-term reconstitution in irradiated adult mice. Blast colony assays revealed that most hemangioblast activity was detected apart from the pre-HSC population, implying the early divergence of pre-HSCs from hemangioblasts. Gene expression profiling suggests that a particular set of transcripts closely associated with adult HSCs is involved in the transition of pre-HSC to embryonic HSCs. We propose an HSC developmental model in which pre-HSCs and embryonic HSCs sequentially give rise to adult types of HSCs in a stepwise manner.
  • K Inamura, T Fujiwara, Y Hoshida, T Isagawa, MH Jones, C Virtanen, M Shimane, Y Satoh, S Okumura, K Nakagawa, E Tsuchiya, S Ishikawa, H Aburatani, H Nomura, Y Ishikawa
    ONCOGENE 24(47) 7105-7113 2005年10月  査読有り
    Current clinical and histopathological criteria used to define lung squamous cell carcinomas (SCCs) are insufficient to predict clinical outcome. To make a clinically useful classification by gene expression profiling, we used a 40 386 element cDNA microarray to analyse 48 SCC, nine adenocarcinoma, and 30 normal lung samples. Initial analysis by hierarchical clustering (HC) allowed division of SCCs into two distinct subclasses. An additional independent round of HC induced a similar partition and consensus clustering with the non-negative matrix factorization approach indicated the robustness of this classification. Kaplan-Meier analysis with the log-rank test pointed to a nonsignificant difference in survival (P = 0.071), but the likelihood of survival to 6 years was significantly different between the two groups (40.5 vs 81.8%, P = 0.014, Z-test). Biological process categories characteristic for each subclass were identified statistically and upregulation of cell-proliferation-related genes was evident in the subclass with poor prognosis. In the subclass with better survival, genes involved in differentiated intracellular functions, such as the MAPKKK cascade, ceramide metabolism, or regulation of transcription, were upregulated. This work represents an important step toward the identification of clinically useful classification for lung SCC.
  • H Mukai, T Isagawa, E Goyama, S Tanaka, NF Bence, A Tamura, Y Ono, RR Kopito
    PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 102(31) 10887-10892 2005年8月  査読有り
    Huntington's disease is a progressive neurodegenerative disorder caused by a polyglutamine repeat expansion in the first exon of the huntingtin (Htt) protein. N-terminal Htt peptides with polyglutamine tracts in the pathological range (51-122 glutamines) form high-molecular-weight protein aggregates with fibrillar morphology in vitro, and they form discrete inclusion bodies in a cell-culture model. However, in some studies, formation of discrete Htt inclusions does not correlate well with cell death. We coexpressed N-terminal Htt fragments containing 91 glutamines fused to different affinity tags in HEK293 cells, and we isolated small aggregates by double sequential-affinity chromatography to assure the isolation of multimeric molecules. Transmission electron microscopy and atomic force microscopy revealed the isolated aggregates as globules or clusters of globules 4-50nm in diameter without any detectable fibrillar species. Because small nonfibrillar oligomers, not mature fibrils, recently have been suggested to be the principal cytotoxic species in neurodegenerative disease, these Htt globular aggregates formed in cells may represent the pathogenic form of mutant Htt.
  • T Isagawa, M Takahashi, T Kato, H Mukai, Y Ono
    MOLECULAR CARCINOGENESIS 43(1) 1-12 2005年5月  査読有り
    PKN1 is a serine/threonine protein kinase that has been reported to mediate cellular response to stress. We show here that in response to arsenite exposure, PKN1 kinase activity was stimulated, which was associated with increased binding of PKN1 to Cdc25C and delayed mitotic entry. A role for PKN1 in mediating arsenite-induced G(2)/M delay was supported by the finding that expression of a constitutively active form of PKN1 (PKN1AF3) in HeLa cells delayed the mitotic entry of cell cycle. Further experiments indicate that PKN1 directly phosphorylated serine 216 (Ser216) in Cdc25C, which then facilitated association between Cdc25C and 14-3-3. Significantly, expression of a phosphorylation mutant of Cdc25C (S21 6A) partially abrogated the cell-cycle arrest in response to arsenite. Together, our results suggest that PKN1 mediates arsenite-induced delay of the G2/M transition by binding to and phoshorylating Cdc25C. (c) 2005 Wiley-Liss, Inc.
  • Y Gotoh, K Oishi, H Shibata, A Yamagiwa, T Isagawa, T Nishimura, E Goyama, M Takahashi, H Mukai, Y Ono
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 314(3) 688-694 2004年2月  査読有り
    PKN1 is a fatty acid and Rho-activated serine/threonine protein kinase whose catalytic domain is highly homologous to protein kinase C (PKC) family. In yeast two-hybrid screening for PKN1 binding proteins, we identified tumor necrosis factor alpha (TNFalpha) receptor-associated factor 2 (TRAF2). TRAF2 is one of the major mediators of TNF receptor superfamily transducing TNF signal to various functional targets, including activation of NF-kappaB, JNK, and apoptosis. FLAG-tagged PKN1 was co-immunoprecipitated with endogenous TRAF2 from HEK293 cell lysate, and in vitro binding assay using the deletion mutants of TRAF2 showed that PKN1 directly binds to the TRAF domain of TRAF2. PKN1 has the TRAF2-binding consensus sequences PXQX (S/T) at amino acid residues 580-584 (PIQES), and P580AQ582A mutant was not co-immunoprecipitated with TRAF2. Furthermore, the reduced expression of PKN1 by RNA interference (RNAi) down-regulated TRAF2-induced NF-kappaB activation in HEK293T cells. These results suggest that PKN1 is involved in TRAF2-NF-kappaB signaling pathway. (C) 2003 Elsevier Inc. All rights reserved.
  • M Takahashi, Y Gotoh, T Isagawa, T Nishimura, E Goyama, HS Kim, H Mukai, Y Ono
    JOURNAL OF BIOCHEMISTRY 133(2) 181-187 2003年2月  査読有り
    PKNalpha is a fatty acid- and Rho-activated serine/threonine protein kinase having a catalytic domain homologous to members of the protein kinase C family. Recently it was reported that PKNalpha is involved in the p38 mitogen-activated protein kinase (MAPK) signaling pathway. To date, however, how PKNalpha regulates the p38gamma MAPK signaling pathway is unclear. Here we demonstrate that PKNalpha efficiently phosphorylates MLTKalpha (MLK-like mitogen-activated protein triple kinase), which was recently identified as a MAPK kinase kinase (MAPKKK) for the p38 MAPK cascade. Phosphorylation of MLTKalpha by PKNalpha enhances its kinase activity in vitro. Expression of the kinase-negative mutant of PKNalpha inhibited the mobility shift of MLTKalpha caused by osmotic shock in SDS-PAGE. Furthermore, PKNalpha associates with each member of the p387gamma MAPK signaling pathway (p38gamma, MKK6, and MLTKalpha). These results suggest that PKNalpha functions as not only an upstream activator of MLTKalpha but also a putative scaffold protein for the p38gamma MAPK signaling pathway.
  • T Taniguchi, T Kawamata, H Mukai, H Hasegawa, T Isagawa, M Yasuda, T Hashimoto, A Terashima, M Nakai, Y Ono, C Tanaka
    JOURNAL OF BIOLOGICAL CHEMISTRY 276(13) 10025-10031 2001年3月  査読有り
    For the phosphorylation state of microtubule-associated protein, tau plays a pivotal role in regulating microtubule networks in neurons. Tau promotes the assembly and stabilization of microtubules, The potential for tau to bind to microtubules is down-regulated after local phosphorylation, When we investigated the effects of PKN activation on tau phosphorylation, we found that PKN triggers disruption of the microtubule array both in vitro and in vivo and predominantly phosphorylates tau in microtubule binding domains (MBDs). PKN has a catalytic domain highly homologous to protein kinase C (PI(C), a kinase that phosphorylates Ser-313 (= Ser-324, the number used in this study) in MBDs, Thus, we identified the phosphorylation sites of PKN and PKC subtypes (PKC-alpha, -betaI, -beta II, -gamma, -delta, -epsilon, -xi, and -lambda) in MBDs, PKN phosphorylates Ser-258, Ser-320, and Ser-352, although all PKC subtypes phosphorylate Ser-258, Ser-293, Ser-324, and Ser-352, There is a PKN-specific phosphorylation site, Ser-320, in MBDs. HIA3, a novel phosphorylation-dependent antibody recognizing phosphorylated tau at Ser-320, showed immunoreactivity in Chinese hamster ovary cells expressing tau and the active form of PKN, but not in Chinese hamster ovary cells expressing tan and the inactive form of PKN, The immunoreactivity for phosphorylated tau at Ser-320 increased in the presence of a phosphatase inhibitor, FK506 treatment, which means that calcineurin (protein phosphatase 2B) may be involved in dephosphorylating tau at Ser-320 site. We also noted that PKN reduces the phosphorylation recognized by the phosphorylation-dependent antibodies AT8, AT180, and AT270 in vivo. Thus PKN serves as a regulator of microtubules by specific phosphorylation of tau, which leads to disruption of tubulin assembly.
  • K Misaki, H Mukai, C Yoshinaga, K Oishi, T Isagawa, M Takahashi, K Ohsumi, T Kishimoto, Y Ono
    PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 98(1) 125-129 2001年1月  査読有り
    The role of PKN, a fatty acid- and Rho small GTPase-activated protein kinase, in cell-cycle regulation was analyzed. Microinjection of the active form of PKN into a Xenopus embryo caused cleavage arrest, whereas normal cell division proceeded in the control embryo microinjected with buffer or the inactive form of PKN. Exogenous addition of the active form of PKN delayed mitotic timing in Xenopus egg cycling extracts judging by morphology of sperm nuclei and Cdc2/cyclin B histone H1 kinase activity. The kinase-negative form of PKN did not affect the timing, suggesting that delayed mitotic timing depends on the kinase activity of PKN. The dephosphorylation of Tyr-15 of Cdc2 was also delayed in correlation with Cdc2/cyclin B histone H1 kinase activation in extracts containing active PKN. The Cdc25C activity for the dephosphorylation of Tyr-15 in Cdc2 was suppressed by pretreatment with the active form of PKN. Furthermore, PKN efficiently phosphorylated Cdc25C in vitro, indicating that PKN directly inhibits Cdc25C activity by phosphorylation. These results suggest that PKN plays a significant role in the control of mitotic timing by inhibition of Cdc25C.
  • M Takahashi, H Mukai, K Oishi, T Isagawa, Y Ono
    JOURNAL OF BIOLOGICAL CHEMISTRY 275(44) 34592-34596 2000年11月  査読有り
    Protein kinase C (PKC) family requires phosphorylation of itself to become competent for responding to second messengers. Much attention has been focused on elucidating the role of phosphorylation in PKC activity; however, it remains unknown where this modification takes place in the cells. This study examines whether anchoring protein is involved in the regulation of PKC phosphorylation. A certain population of PKC epsilon in rat brain extracts as well as that expressed in COS7 cells was associated with an endogenous anchoring protein CG-NAP (centrosome and Golgi localized PKN-associated protein). Pulse chase experiments revealed that the associated PKC epsilon was an immature species at the hypophosphorylated state. In vitro binding studies confirmed that non- or hypophosphorylated PKC epsilon directly bound to CG-NAP via its catalytic domain, whereas sufficiently phosphorylated PKC epsilon did not. PKC epsilon mutant at a potential phosphorylation site of Thr-566 or Ser-729 to Ala, possessing almost no catalytic activity, was associated and co-localized with CG-NAP at Golgi/centrosome area. On the other hand, wild type and a phosphorylation-mimicking mutant at Thr-566 were mainly distributed in cytosol and represented second messenger-dependent catalytic activation. These results suggest that CG-NAP anchors hypophosphorylated PKC epsilon at the Golgi/centrosome area during maturation and serves as a scaffold for the phosphorylation reaction.
  • T Isagawa, H Mukai, K Oishi, T Taniguchi, H Hasegawa, T Kawamata, C Tanaka, Y Ono
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 273(1) 209-212 2000年6月  査読有り
    We analyzed the effects of PKN alpha and protein kinase C (PKC) on phosphorylation of tau protein by glycogen synthase kinase (GSK)-3 beta using monoclonal antibodies (AT8, AT180, and AT270), These antibodies are highly specific for phosphorylated tan in Alzheimer paired helical filaments, and recognize phosphorylated Ser202/Thr205, Thr231, and Thr181 of tan protein, respectively, Immunoblot analysis demonstrated that PKN alpha and PKC did not directly phosphorylate their sites, whereas GSK-3 beta efficiently did so. Incubating GSK-3 beta with PKN alpha or PKC subtypes inhibited subsequent GSK-3 beta-induced AT8 and AT270 immunoreactivity. However, the constitutive active form of the GSK-3 beta(S9A) mutant was almost totally inert to each enzyme. Incubating tau with PKNa increased the GSK-3 beta-induced AT180 immunoreactivity, which was further enhanced when the S9A mutant was used instead of the wild type GSK-3 beta. These results suggest that PKN alpha and PKC directly inhibit GSK-3 beta activity at least in part by phosphorylating Ser9 of GSK-3 beta, and that they indirectly suppress GSK-3 beta-stimulated phosphorylation of tau at amino acids Ser202/Thr205 and Thr181, but enhanced phosphorylation at Thr231 through phosphorylation at other sites of tau. (C) 2000 Academic Press.

MISC

 81

共同研究・競争的資金等の研究課題

 11

産業財産権

 1