Network


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

Hotspot


Dive into the research topics where Takahiro Maeda is active.

Publication


Featured researches published by Takahiro Maeda.


Nature | 2005

Role of the proto-oncogene Pokemon in cellular transformation and ARF repression

Takahiro Maeda; Robin M. Hobbs; Taha Merghoub; Ilhem Guernah; Arthur Zelent; Carlos Cordon-Cardo; Julie Teruya-Feldstein; Pier Paolo Pandolfi

Aberrant transcriptional repression through chromatin remodelling and histone deacetylation has been postulated to represent a driving force underlying tumorigenesis because histone deacetylase inhibitors have been found to be effective in cancer treatment. However, the molecular mechanisms by which transcriptional derepression would be linked to tumour suppression are poorly understood. Here we identify the transcriptional repressor Pokemon (encoded by the Zbtb7 gene) as a critical factor in oncogenesis. Mouse embryonic fibroblasts lacking Zbtb7 are completely refractory to oncogene-mediated cellular transformation. Conversely, Pokemon overexpression leads to overt oncogenic transformation both in vitro and in vivo in transgenic mice. Pokemon can specifically repress the transcription of the tumour suppressor gene ARF through direct binding. We find that Pokemon is aberrantly overexpressed in human cancers and that its expression levels predict biological behaviour and clinical outcome. Pokemons critical role in cellular transformation makes it an attractive target for therapeutic intervention.


Cancer Cell | 2012

Altered Microenvironmental Regulation of Leukemic and Normal Stem Cells in Chronic Myelogenous Leukemia

Bin Zhang; Yin Wei Ho; Qin Huang; Takahiro Maeda; Allen Lin; Sung Uk Lee; Alan Hair; Tessa L. Holyoake; Claudia S. Huettner; Ravi Bhatia

We characterized leukemia stem cells (LSC) in chronic phase chronic myelogenous leukemia (CML) using a transgenic mouse model. LSC were restricted to cells with long-term hematopoietic stem cell (LTHSC) phenotype. CML LTHSC demonstrated reduced homing and retention in the bone marrow (BM), related to decreased CXCL12 expression in CML BM, resulting from increased G-CSF production by leukemia cells. Altered cytokine expression in CML BM was associated with selective impairment of normal LTHSC growth and a growth advantage to CML LTHSC. Imatinib (IM) treatment partially corrected abnormalities in cytokine levels and LTHSC growth. These results were validated using human CML samples and provide improved understanding of microenvironmental regulation of normal and leukemic LTHSC and their response to IM in CML.


Journal of Biological Chemistry | 2014

Characterization of Genomic Deletion Efficiency Mediated by Clustered Regularly Interspaced Palindromic Repeats (CRISPR)/Cas9 Nuclease System in Mammalian Cells

Matthew C. Canver; Daniel E. Bauer; Abhishek Dass; Yvette Y. Yien; Jacky Chung; Takeshi Masuda; Takahiro Maeda; Barry H. Paw; Stuart H. Orkin

Background: CRISPR/Cas9-directed cleavages may result in genomic deletion. Results: CRISPR/Cas9-produced genomic deletion frequency is inversely related to deletion size, with large deletions and inversions practicable and biallelic deletions exceeding probabilistic expectation. Conclusion: Biallelic, large genomic deletions are efficiently engineered in mammalian cells by CRISPR/Cas9. Significance: CRISPR/Cas9-mediated genomic deletion represents a robust method for loss-of-function studies in mammalian cells. The clustered regularly interspaced palindromic repeats (CRISPR)/CRISPR-associated (Cas) 9 nuclease system has provided a powerful tool for genome engineering. Double strand breaks may trigger nonhomologous end joining repair, leading to frameshift mutations, or homology-directed repair using an extrachromosomal template. Alternatively, genomic deletions may be produced by a pair of double strand breaks. The efficiency of CRISPR/Cas9-mediated genomic deletions has not been systematically explored. Here, we present a methodology for the production of deletions in mammalian cells, ranging from 1.3 kb to greater than 1 Mb. We observed a high frequency of intended genomic deletions. Nondeleted alleles are nonetheless often edited with inversions or small insertion/deletions produced at CRISPR recognition sites. Deleted alleles also typically include small insertion/deletions at predicted deletion junctions. We retrieved cells with biallelic deletion at a frequency exceeding that of probabilistic expectation. We demonstrate an inverse relationship between deletion frequency and deletion size. This work suggests that CRISPR/Cas9 is a robust system to produce a spectrum of genomic deletions to allow investigation of genes and genetic elements.


Nature Neuroscience | 2015

Central role for PICALM in amyloid-β blood-brain barrier transcytosis and clearance

Zhen Zhao; Abhay P. Sagare; Qingyi Ma; Matthew R. Halliday; Pan Kong; Kassandra Kisler; Ethan A. Winkler; Anita Ramanathan; Takahisa Kanekiyo; Guojun Bu; Nelly Chuqui Owens; Sanket V Rege; Gabriel Si; Ashim Ahuja; Donghui Zhu; Carol A. Miller; Julie A. Schneider; Manami Maeda; Takahiro Maeda; Tohru Sugawara; Justin K. Ichida; Berislav V. Zlokovic

PICALM is a highly validated genetic risk factor for Alzheimers disease (AD). We found that reduced expression of PICALM in AD and murine brain endothelium correlated with amyloid-β (Aβ) pathology and cognitive impairment. Moreover, Picalm deficiency diminished Aβ clearance across the murine blood-brain barrier (BBB) and accelerated Aβ pathology in a manner that was reversible by endothelial PICALM re-expression. Using human brain endothelial monolayers, we found that PICALM regulated PICALM/clathrin-dependent internalization of Aβ bound to the low density lipoprotein receptor related protein-1, a key Aβ clearance receptor, and guided Aβ trafficking to Rab5 and Rab11, leading to Aβ endothelial transcytosis and clearance. PICALM levels and Aβ clearance were reduced in AD-derived endothelial monolayers, which was reversible by adenoviral-mediated PICALM transfer. Inducible pluripotent stem cell–derived human endothelial cells carrying the rs3851179 protective allele exhibited higher PICALM levels and enhanced Aβ clearance. Thus, PICALM regulates Aβ BBB transcytosis and clearance, which has implications for Aβ brain homeostasis and clearance therapy.


Cancer Research | 2005

The Transcription Factor Pokemon: A New Key Player in Cancer Pathogenesis

Takahiro Maeda; Robin M. Hobbs; Pier Paolo Pandolfi

Learning how critical cell regulatory pathways are controlled may lead to new opportunities for cancer treatment. We recently identified the transcription factor Pokemon as a central regulator of the important tumor suppressor ARF. Pokemon is overexpressed in multiple human cancers and cells lacking Pokemon are refractory to oncogenic transformation. These findings suggest that Pokemon may offer an effective new target for cancer therapeutics.


International Journal of Hematology | 2005

A novel plasmacytoid dendritic cell line, CAL-1, established from a patient with blastic natural killer cell lymphoma.

Takahiro Maeda; Kenn Murata; Takuya Fukushima; Kazuyuki Sugahara; Kazuto Tsuruda; Masanobu Anami; Yasuyuki Onimaru; Kunihiro Tsukasaki; Masao Tomonaga; Ryouzou Moriuchi; Hiroo Hasegawa; Yasuaki Yamada; Shimeru Kamihira

Blastic natural killer (NK) cell lymphoma corresponding to CD4+CD56+ malignancies is a novel disease entity, according to the results of clinical, morphologic, and immunologic studies. It is especially noteworthy that this disease likely arises from plasmacytoid dendritic cells (pDCs), described previously as plasmacytoid T-cells, which have an important role in innate and adaptive immunity. However, the exact relationship between the tumor cells and pDCs remains to be elucidated.We encountered a patient with typical blastic NK cell lymphoma, which later converted to leukemic manifestations, and tried to establish a cell line using the leukemic cells.We succeeded in establishment of a novel cell line,CAL-1, which originated from the primary malignant cells.The genetic and phenotypic features of CAL-1 cells bear a similarity to those of pDCs, namely, plasmacytoid morphology at light and electron microscopy; negative results for CD11c and lineage-associated markers of CD3, CD14, CD19, and CD16; positive results for HLA-DR, CD4, CD56, CD45RA, and CD123; and negative results for TCR and IgH gene rearrangements. An interesting finding was that CAL-1 cells change morphologically into the mature DC appearance with many long dendrites after short-term culture in the presence of granulocyte-macrophage colony-stimulating factor and interleukin 3.CAL-1 cells can secrete tumor necrosis factorαbut not interferon α.Thus although they do not share in part phenotypic and functional features with their normal counterparts, CAL-1 cells mostly exhibit a striking pDC phenotype.We describe the first novel pDC cell line of CAL-1.This cell line should open the opportunity for study not only of CD4+CD56+ tumor cells but also of pDCs in vitro.


Developmental Cell | 2009

LRF Is an Essential Downstream Target of GATA1 in Erythroid Development and Regulates BIM-Dependent Apoptosis

Takahiro Maeda; Keisuke Ito; Taha Merghoub; Laura Poliseno; Robin M. Hobbs; Guocan Wang; Lin Dong; Manami Maeda; Louis C. Dore; Arthur Zelent; Lucio Luzzatto; Julie Teruya-Feldstein; Mitchell J. Weiss; Pier Paolo Pandolfi

GATA-1-dependent transcription is essential for erythroid differentiation and maturation. Suppression of programmed cell death is also thought to be critical for this process; however, the link between these two features of erythropoiesis has remained elusive. Here, we show that the POZ-Krüppel family transcription factor, LRF (also known as Zbtb7a/Pokemon), is a direct target of GATA1 and plays an essential antiapoptotic role during terminal erythroid differentiation. We find that loss of Lrf leads to lethal anemia in embryos, due to increased apoptosis of late-stage erythroblasts. This programmed cell death is Arf and p53 independent and is instead mediated by upregulation of the proapoptotic factor Bim. We identify Lrf as a direct repressor of Bim transcription. In strong support of this mechanism, genetic Bim loss delays the lethality of Lrf-deficient embryos and rescues their anemia phenotype. Thus, our data define a key transcriptional cascade for effective erythropoiesis, whereby GATA-1 suppresses BIM-mediated apoptosis via LRF.


Science | 2016

Transcription factors LRF and BCL11A independently repress expression of fetal hemoglobin

Takeshi Masuda; Xin Wang; Manami Maeda; Matthew C. Canver; Falak Sher; Alister P. W. Funnell; Chris Fisher; Maria Suciu; Gabriella E. Martyn; Laura J. Norton; Catherine Zhu; Ryo Kurita; Yukio Nakamura; Jian Xu; Douglas R. Higgs; Merlin Crossley; Daniel E. Bauer; Stuart H. Orkin; Peter V. Kharchenko; Takahiro Maeda

Reactivating the fetal globin gene Mutation of adult-type globin genes causes sickle cell disease and thalassemia. Although treating these hemoglobinopathies with gene therapy is possible, there is a pressing need for pharmacologic approaches to treat general patient populations. One promising approach is to reactivate repressed expression of fetal-type hemoglobin (HbF) in adult erythroid cells. Masuda et al. reveal a molecular mechanism governing HbF repression as mediated by the LRF/ZBTB7A transcription factor. The study may encourage the development of new HbF reactivation therapies for hemoglobinopathies. Science, this issue p. 285 Reactivation of fetal globin gene expression may enable treatment of hemoglobinopathies. Genes encoding human β-type globin undergo a developmental switch from embryonic to fetal to adult-type expression. Mutations in the adult form cause inherited hemoglobinopathies or globin disorders, including sickle cell disease and thalassemia. Some experimental results have suggested that these diseases could be treated by induction of fetal-type hemoglobin (HbF). However, the mechanisms that repress HbF in adults remain unclear. We found that the LRF/ZBTB7A transcription factor occupies fetal γ-globin genes and maintains the nucleosome density necessary for γ-globin gene silencing in adults, and that LRF confers its repressive activity through a NuRD repressor complex independent of the fetal globin repressor BCL11A. Our study may provide additional opportunities for therapeutic targeting in the treatment of hemoglobinopathies.


Immunological Reviews | 2012

POK/ZBTB proteins: an emerging family of proteins that regulate lymphoid development and function

Sung Uk Lee; Takahiro Maeda

Summary:u2002 The germinal center (GC) is a unique histological structure found in peripheral lymphoid organs. GCs provide an important source of humoral immunity by generating high affinity antibodies against a pathogen. The GC response is tightly regulated during clonal expansion, immunoglobulin modification, and affinity maturation, whereas its deregulation has a detrimental effect on immune function, leading to development of diseases, such as lymphoma and autoimmunity. LRF (lymphoma/leukemia‐related factor), encoded by the ZBTB7A gene, is a transcriptional repressor belonging to the POK (POZ and Krüppel)/ZBTB (zing finger and BTB) protein family. LRF was originally identified as a PLZF (promyelocytic leukemia zinc finger) homolog that physically interacts with BCL6 (B‐cell lymphoma 6), whose expression is required for GC formation and associated with non‐Hodgkin’s lymphoma. Recently, our group demonstrated that LRF plays critical roles in regulating lymphoid lineage commitment, mature B‐cell development, and the GC response via distinct mechanisms. Herein, we review POK/ZBTB protein function in lymphoid development, with particular emphasis on the role of LRF in GC B cells.


Nature Communications | 2015

Notch signal strength controls cell fate in the haemogenic endothelium.

Leonor Gama-Norton; Eva Ferrando; Cristina Ruiz-Herguido; Zhenyi Liu; Jordi Guiu; Abul B.M.M.K. Islam; Sung-Uk Lee; Minhong Yan; Cynthia J. Guidos; Nuria Lopez-Bigas; Takahiro Maeda; Lluis Espinosa; Raphael Kopan; Anna Bigas

Acquisition of the arterial and haemogenic endothelium fates concurrently occur in the aorta–gonad–mesonephros (AGM) region prior to haematopoietic stem cell (HSC) generation. The arterial programme depends on Dll4 and the haemogenic endothelium/HSC on Jag1-mediated Notch1 signalling. How Notch1 distinguishes and executes these different programmes in response to particular ligands is poorly understood. By using two Notch1 activation trap mouse models with different sensitivity, here we show that arterial endothelial cells and HSCs originate from distinct precursors, characterized by different Notch1 signal strengths. Microarray analysis on AGM subpopulations demonstrates that the Jag1 ligand stimulates low Notch strength, inhibits the endothelial programme and is permissive for HSC specification. In the absence of Jag1, endothelial cells experience high Dll4-induced Notch activity and select the endothelial programme, thus precluding HSC formation. Interference with the Dll4 signal by ligand-specific blocking antibodies is sufficient to inhibit the endothelial programme and favour specification of the haematopoietic lineage.

Collaboration


Dive into the Takahiro Maeda's collaboration.

Top Co-Authors

Avatar

Manami Maeda

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sung-Uk Lee

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Julie Teruya-Feldstein

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Pier Paolo Pandolfi

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nagisa Sakurai

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robin M. Hobbs

Australian Regenerative Medicine Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge