Network


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

Hotspot


Dive into the research topics where Tsukasa Okuda is active.

Publication


Featured researches published by Tsukasa Okuda.


Cell | 1996

AML1, the target of multiple chromosomal translocations in human leukemia, is essential for normal fetal liver hematopoiesis.

Tsukasa Okuda; Jan van Deursen; Scott W. Hiebert; Gerard Grosveld; James R. Downing

The AML1-CBF beta transcription factor is the most frequent target of chromosomal rearrangements in human leukemia. To investigate its normal function, we generated mice lacking AML1. Embryos with homozygous mutations in AML1 showed normal morphogenesis and yolk sac-derived erythropoiesis, but lacked fetal liver hematopoiesis and died around E12.5. Sequentially targeted AML1-/-es cell retained their capacity to differentiate into primitive erythroid cells in vitro; however, no myeloid or erythroid progenitors of definitive hematopoietic origin were detected in either the yolk sac or fetal livers of mutant embryos. Moreover, this hematopoietic defect was intrinsic to the stem cells in that AML1-/-ES cells failed to contribute to hematopoiesis in chimeric animals. These results suggest that AML1-regulated target genes are essential for definitive hematopoiesis of all lineages.


Leukemia | 1997

Internal tandem duplication of the FLT3 gene is preferentially seen in acute myeloid leukemia and myelodysplastic syndrome among various hematological malignancies. A study on a large series of patients and cell lines.

Shouhei Yokota; Hitoshi Kiyoi; Mitsushige Nakao; Toshiki Iwai; Shinichi Misawa; Tsukasa Okuda; Yoshiaki Sonoda; Tatsuo Abe; Kahsima K; Matsuo Y; Tomoki Naoe

In this study, we examined a large number of patients to clarify the distribution and frequency of a recently described FLT3 tandem duplication among hematopoietic malignancies, including 112 acute myelocytic leukemia (AML), 55 acute lymphoblastic leukemia (ALL), 37 myelodysplastic syndrome (MDS), 20 chronic myelogenous leukemia (CML), 30 non-Hodgkin’s lymphoma (NHL), 14 adult T cell leukemia, 15 chronic lymphocytic leukemia (CLL) and 38 multiple myeloma (MM). We also evaluated 71 cell lines derived from 11 AML, 31 ALL, two hairy cell leukemia, three acute unclassified leukemia, 10 CML, 12 NHL including six Burkitt’s lymphoma, and two MM. Using genomic PCR of exon 11 coding for the juxtamembrane (JM) domain and first amino acids of the 5′-tyrosine kinase (TK) domain, this length mutation was found only in AML (22/112, 20%) and MDS (1/37). According to the FAB subclassification, they were 5/18 (28%) of M1, 4/29 (14%) of M2, 3/17 (18%) of M3, 6/24 (25%) of M4, 4/20 (20%) of M5 and 1/9 of refractory anemia with excess of blast in transformation. In the various cell lines examined, this abnormality was determined in only one derived from AML and never found in other hematological malignancies. The sequence analysis of the abnormal PCR products revealed that 23 of 24 showed internal tandem duplication with or without insertion of nucleotides. In one AML, insertion and deletion without duplication was determined. All 24 lengthened sequences were in-frame. Duplication takes place in the sequence coding for the JM domain and leaves the TK domain intact. In conclusion, we emphasize that the length mutation of FLT3 at JM/TK-I domains were restricted to AML and MDS. Since all these mutations resulted in in-frame, this abnormality might function for the proliferation of leukemic cells.


Science | 2008

Repression of the Transcription Factor Th-POK by Runx Complexes in Cytotoxic T Cell Development

Ruka Setoguchi; Masashi Tachibana; Yoshinori Naoe; Sawako Muroi; Kaori Akiyama; Chieko Tezuka; Tsukasa Okuda; Ichiro Taniuchi

Mouse CD4+CD8+ double-positive (DP) thymocytes differentiate into CD4+ helper-lineage cells upon expression of the transcription factor Th-POK but commit to the CD8+ cytotoxic lineage in its absence. We report the redirected differentiation of class I–restricted thymocytes into CD4+CD8– helper-like T cells upon loss of Runx transcription factor complexes. A Runx-binding sequence within the Th-POK locus acts as a transcriptional silencer that is essential for Th-POK repression and for development of CD8+ T cells. Thus, Th-POK expression and genetic programming for T helper cell development are actively inhibited by Runx-dependent silencer activity, allowing for cytotoxic T cell differentiation. Identification of the transcription factors network in CD4 and CD8 lineage choice provides insight into how distinct T cell subsets are developed for regulating the adaptive immune system.


Molecular and Cellular Biology | 2000

Biological characteristics of the leukemia-associated transcriptional factor AML1 disclosed by hematopoietic rescue of AML1-deficient embryonic stem cells by using a knock-in strategy.

Tsukasa Okuda; Kiyoshi Takeda; Yasuko Fujita; Motohiro Nishimura; Shigeki Yagyu; Makie Yoshida; Shizuo Akira; James R. Downing; Tatsuo Abe

ABSTRACT AML1 is one of the most frequently mutated genes associated with human acute leukemia and encodes the DNA-binding subunit of the heterodimering transcriptional factor complex, core-binding factor (CBF) (or polyoma enhancer binding protein 2 [PEBP2]). A null mutation in either AML1 or its dimerizing partner, CBFβ, results in embryonic lethality secondary to a complete block in fetal liver hematopoiesis, indicating an essential role of this transcription complex in the development of definitive hematopoiesis. The hematopoietic phenotype that results from the loss of AML1 can be replicated in vitro with a two-step culture system of murine embryonic stem (ES) cells. Using this experimental system, we now demonstrate that this hematopoietic defect can be rescued by expressing thePEBP2αB1 (AML1b) isoform under the endogenousAML1-regulatory sequences through a knock-in (targeted insertion) approach. Moreover, we demonstrate that the rescuedAML1 −/− ES cell clones contribute to lymphohematopoiesis within the context of chimeric animals. Rescue requires the transcription activation domain of AML1 but does not require the C-terminal VWRPY motif, which is conserved in all AML1 family members and has been shown to interact with the transcriptional corepressor, Groucho/transducin-like Enhancer of split. Taken together, these data provide compelling evidence that the phenotype seen inAML1-deficient mice is due solely to the loss of transcriptionally active AML1.


International Journal of Hematology | 2001

RUNX1/AML1: A Central Player in Hematopoiesis

Tsukasa Okuda; Motohiro Nishimura; Mitsushige Nakao; Yasuko Fujitaa

It has been well established that a number of transcription factors play critical roles in regulating the fate of hematopoietic stem cell populations. One of them is the leukemia-associated transcription factor acute myeloid leukemia 1 (AML1; also known as runt-related transcription factor 1, or RUNX1). This gene was originally cloned from the breakpoint of the t(8;21) reciprocal chromosome translocation and was later recognized as one of the most frequent targets of leukemia-associated gene aberrations. Gene-targeting experiments revealed that transcriptionally active AML1 is essential for the establishment of definitive hematopoiesis. More specifically, this gene functions in the emergence of the hematopoietic progenitor cells from the hemogenic endothelium by budding in the aorta-gonad-mesonephros region, and its expression points to the sites with strong potential for the emergence of hematopoietic stem cells. This review discusses aspects of the biologic properties of AML1 in early hematopoietic development.


The Journal of Neuroscience | 2008

Mechanisms of Compartmentalized Expression of Mrg Class G-Protein-Coupled Sensory Receptors

Yang Liu; Fu Chia Yang; Tsukasa Okuda; Xinzhong Dong; Mark J. Zylka; Chih Li Chen; David J. Anderson; Rohini Kuner; Qiufu Ma

Mrg class G-protein-coupled receptors (GPCRs) are expressed exclusively in sensory neurons in the trigeminal and dorsal root ganglia. Pharmacological activation of Mrg proteins is capable of modulating sensory neuron activities and elicits nociceptive effects. In this study, we illustrate a control mechanism that allows the Runx1 runt domain transcription factor to generate compartmentalized expression of these sensory GPCRs. Expression of MrgA, MrgB, and MrgC subclasses is confined to an “A/B/C” neuronal compartment that expresses Runx1 transiently (or does not express Runx1), whereas MrgD expression is restricted to a “D” compartment with persistent Runx1 expression. Runx1 is initially required for the expression of all Mrg genes. However, during late development Runx1 becomes a repressor for MrgA/B/C genes. As a result, MrgA/B/C expression persists only in the Runx1− “A/B/C” compartment. In Δ446 mice, in which Runx1 lacks the C-terminal repression domain, expression of MrgA/B/C genes is dramatically expanded into the Runx1+ “D” compartment. MrgD expression, however, is resistant to Runx1-mediated repression in the “D” compartment. Therefore, the creation of Runx1+ and Runx1− compartments, in conjunction with different responses of Mrg genes to Runx1-mediated repression, results in the compartmentalized expression of MrgA/B/C versus MrgD genes. Within the MrgA/B/C compartment, MrgB4-expressing neurons innervate exclusively the hairy skin. Here we found that Smad4, a downstream component of bone morphological protein-mediated signaling, is required selectively for the expression of MrgB4. Our study suggests a new line of evidence that specification of sensory subtypes is established progressively during perinatal and postnatal development.


Cancer | 1990

The unbalanced 1;7 translocation in de novo myelodysplastic syndrome and its clinical implication

Shigeo Horiike; Masafumi Taniwaki; Shinichi Misawa; Hikari Nishigaki; Tsukasa Okuda; Shohei Yokota; Kei Kashima; Johji Inazawa; Tatsuo Abe

In our chromosome study of 97 patients with myelodysplastic syndrome (MDS), six showed an unbalanced translocation between chromosomes 1 and 7 [−7, +der(1)t(1;7)(p11;p11)]. All of them had morphologic myelodysplasia in trilineage of bone marrow cells, and cytopenia was the major finding in the peripheral blood. All six patients had symptoms of infection at the time of diagnosis, and five showed immunologic abnormalities (polyclonal hypergammaglobulinemia in four and increased marrow plasma cells in three). None of the patients survived more than 11 months after the diagnosis; the median survival time was 4 months. Both of the two patients whose karyotypes were reexamined in the course of their disease showed karyotypic evolution accompanying the coincidental leukemic transformation. Six patients with MDS who had the same chromosome abnormality [t(1;7)] are described and their characteristic clinical features are presented.


Journal of Biological Chemistry | 2008

AML1/RUNX1 Works as a Negative Regulator of c-Mpl in Hematopoietic Stem Cells

Yusuke Satoh; Itaru Matsumura; Hirokazu Tanaka; Sachiko Ezoe; Kentaro Fukushima; Masahiro Tokunaga; Masato Yasumi; Hirohiko Shibayama; Masao Mizuki; Takumi Era; Tsukasa Okuda; Yuzuru Kanakura

In this study, we analyzed the roles for AML1/RUNX1 in the regulation of the c-mpl promoter. Wild-type AML1 activated the c-mpl promoter through the proximal AML-binding site in luciferase assays using 293T and HeLa cells. In accord with this result, electrophoretic mobility shift assay and chromatin immunoprecipitation assays demonstrated that AML1 bound to this site. Next, we analyzed the function of AML1 using a mutant of AML1 lacking the C terminus (AML1dC), which was originally found in a patient with myelodysplastic syndromes. AML1dC dominant-negatively suppressed transcriptional activity of wild-type AML1. However, unexpectedly, AML1dC-transduced murine c-Kit+Sca1+Lineage- cells expressed c-mpl mRNA and c-Mpl protein more abundantly than mock-transduced cells, which led to the enhanced thrombopoietin-mediated proliferation. Moreover, when AML1dC was induced to express during the development of hematopoietic cells from embryonic stem (ES) cells, AML1dC augmented the c-Mpl expression on hematopoietic stem/progenitor cells. Furthermore, we found that early hematopoietic cells that derived from AML1+/- ES cells expressed c-Mpl more intensely than those that developed from wild-type ES cells. In contrast, AML1dC hardly affected c-Mpl expression and maturation of megakaryocytes. As for the mechanism of the different roles of AML1 in the regulation of the c-mpl promoter, we found that AML1 forms a complex with a transcription repressor mSin3A on the c-mpl promoter in hematopoietic stem/progenitor cells, although it forms a complex with a transcription activator p300 on the same promoter in megakaryocytic cells. Together, these data indicate that AML1 can regulate the c-mpl promoter both positively and negatively by changing the binding partner according to cell types.


British Journal of Haematology | 2004

Novel loss-of-function mutations of the haematopoiesis-related transcription factor, acute myeloid leukaemia 1/runt-related transcription factor 1, detected in acute myeloblastic leukaemia and myelodysplastic syndrome.

Mitsushige Nakao; Shigeo Horiike; Yoko Fukushima-Nakase; Motohiro Nishimura; Yasuko Fujita; Masafumi Taniwaki; Tsukasa Okuda

AML1/RUNX1, which encodes a transcription factor essential for definitive haematopoiesis, is a frequent target of leukaemia‐associated chromosome translocations. Point mutations of this gene have also recently been associated with leukaemia and myelodysplastic syndrome (MDS). To further define the frequency and biological characteristics of AML1 mutations, we have examined 170 cases of such diseases. Mutations within the runt‐domain were identified in five cases: one of de novo acute myeloid leukaemia (AML) and four of MDS. Where multiple time point samples were available, mutations were detected in the earliest samples, which persisted throughout the disease course. Of the five mutations, one was a silent mutation, two were apparent loss‐of‐function mutations caused by N‐terminal truncation, and two were insertions, I150ins and K168ins, which preserved most of the AML1 DNA‐binding domain. Both AML1 molecules with insertion mutations were non‐functional in that they were unable to rescue haematological defects in AML1‐deficient mouse embryonic stem cells. In addition, activating mutations of N‐ras, deletion of chromosome 12p, or inactivation of TP53 accompanied some of the AML1 mutations. Together, these observations strongly suggest that one‐allele inactivation of AML1 serves as an initial or early event that plays an important role in the eventual development of overt diseases with additional genetic alterations.


Leukemia Research | 1988

Detection of karyotypic abnormalities in most patients with acute nonlymphocytic leukemia by adding ethidium bromide to short-term cultures

Shinichi Misawa; Hiromi Yashige; Shigeo Horiike; Masafumi Taniwaki; Hikari Nishigaki; Tsukasa Okuda; Shouhei Yokota; Shoichiro Tsuda; J. Edagawa; Hitoshi Imanishi; Tatsuro Takino; Johji Inazawa; Tatsuo Abe; Shinobu Nakanishi; Masao Nakagawa; Hiroshi Kobayashi; Taira Maekawa; Hiroshi Fujii; Teruaki Akaogi; Hideo Hayashi; Yoshihide Fujiyama; Masatoshi Kohsaki

A modified short-term culture method, in which cultured bone marrow cells were treated with ethidium bromide to prevent chromosome condensation was used to study the chromosomes of 70 patients with acute nonlymphocytic leukemia. Clonal karyotypic abnormalities were detected in 60 patients. Among these, 35 patients showed one of recurrent type specific alterations. A close relationship between karyotypes and clinical outcome was shown: thus, t(8;21) or a single miscellaneous chromosomal defect associated with a favourable prognosis whereas t(9;11) or a complex karyotype related to a poor prognosis. The ten cytogenetically normal patients did not appear to have a favourable prognosis.

Collaboration


Dive into the Tsukasa Okuda's collaboration.

Top Co-Authors

Avatar

Masafumi Taniwaki

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Shinichi Misawa

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Tatsuo Abe

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hikari Nishigaki

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Shigeo Horiike

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Shoichiro Tsuda

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Tatsuro Takino

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

James R. Downing

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Johji Inazawa

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Shouhei Yokota

Kyoto Prefectural University of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge