Network


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

Hotspot


Dive into the research topics where Manami Maeda is active.

Publication


Featured researches published by Manami Maeda.


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.


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.


Journal of Clinical Investigation | 2011

The LRF transcription factor regulates mature B cell development and the germinal center response in mice

Nagisa Sakurai; Manami Maeda; Sung Uk Lee; Yuichi Ishikawa; Min Li; John C. Williams; Lisheng Wang; Leila Su; Mai Suzuki; Toshiki Saito; Shigeru Chiba; Stefano Casola; Hideo Yagita; Julie Teruya-Feldstein; Shinobu Tsuzuki; Ravi Bhatia; Takahiro Maeda

B cells play a central role in immune system function. Deregulation of normal B cell maturation can lead to the development of autoimmune syndromes as well as B cell malignancies. Elucidation of the molecular features of normal B cell development is important for the development of new target therapies for autoimmune diseases and B cell malignancies. Employing B cell-specific conditional knockout mice, we have demonstrated here that the transcription factor leukemia/lymphoma-related factor (LRF) forms an obligate dimer in B cells and regulates mature B cell lineage fate and humoral immune responses via distinctive mechanisms. Moreover, LRF inactivation in transformed B cells attenuated their growth rate. These studies identify what we believe to be a new key factor for mature B cell development and provide a rationale for targeting LRF dimers for the treatment of autoimmune diseases and B cell malignancies.


Blood | 2013

LRF-mediated Dll4 repression in erythroblasts is necessary for hematopoietic stem cell maintenance

Sung Uk Lee; Manami Maeda; Yuichi Ishikawa; Sierra Min Li; Anne Wilson; Adrian M. Jubb; Nagisa Sakurai; Lihong Weng; Emma Fiorini; Freddy Radtke; Minhong Yan; H. Robson MacDonald; Ching Cheng Chen; Takahiro Maeda

Hematopoietic stem cells (HSCs) are the most primitive cells in the hematopoietic system and are under tight regulation for self-renewal and differentiation. Notch signals are essential for the emergence of definitive hematopoiesis in mouse embryos and are critical regulators of lymphoid lineage fate determination. However, it remains unclear how Notch regulates the balance between HSC self-renewal and differentiation in the adult bone marrow (BM). Here we report a novel mechanism that prevents HSCs from undergoing premature lymphoid differentiation in BM. Using a series of in vivo mouse models and functional HSC assays, we show that leukemia/lymphoma related factor (LRF) is necessary for HSC maintenance by functioning as an erythroid-specific repressor of Delta-like 4 (Dll4) expression. Lrf deletion in erythroblasts promoted up-regulation of Dll4 in erythroblasts, sensitizing HSCs to T-cell instructive signals in the BM. Our study reveals novel cross-talk between HSCs and erythroblasts, and sheds a new light on the regulatory mechanisms regulating the balance between HSC self-renewal and differentiation.


Haematologica | 2015

Role of the clathrin adaptor PICALM in normal hematopoiesis and polycythemia vera pathophysiology

Yuichi Ishikawa; Manami Maeda; Mithun Pasham; François Aguet; Silvia K. Tacheva-Grigorova; Takeshi Masuda; Hai Yi; Sung Uk Lee; Jian Xu; Julie Teruya-Feldstein; Maria Ericsson; Ann Mullally; John E. Heuser; Tom Kirchhausen; Takahiro Maeda

Clathrin-dependent endocytosis is an essential cellular process shared by all cell types. Despite this, precisely how endocytosis is regulated in a cell-type-specific manner and how this key pathway functions physiologically or pathophysiologically remain largely unknown. PICALM, which encodes the clathrin adaptor protein PICALM, was originally identified as a component of the CALM/AF10 leukemia oncogene. Here we show, by employing a series of conditional Picalm knockout mice, that PICALM critically regulates transferrin uptake in erythroid cells by functioning as a cell-type-specific regulator of transferrin receptor endocytosis. While transferrin receptor is essential for the development of all hematopoietic lineages, Picalm was dispensable for myeloid and B-lymphoid development. Furthermore, global Picalm inactivation in adult mice did not cause gross defects in mouse fitness, except for anemia and a coat color change. Freeze-etch electron microscopy of primary erythroblasts and live-cell imaging of murine embryonic fibroblasts revealed that Picalm function is required for efficient clathrin coat maturation. We showed that the PICALM PIP2 binding domain is necessary for transferrin receptor endocytosis in erythroblasts and absolutely essential for erythroid development from mouse hematopoietic stem/progenitor cells in an erythroid culture system. We further showed that Picalm deletion entirely abrogated the disease phenotype in a Jak2V617F knock-in murine model of polycythemia vera. Our findings provide new insights into the regulation of cell-type-specific transferrin receptor endocytosis in vivo. They also suggest a new strategy to block cellular uptake of transferrin-bound iron, with therapeutic potential for disorders characterized by inappropriate red blood cell production, such as polycythemia vera.


eLife | 2017

Erythropoietin signaling regulates heme biosynthesis

Jacky Chung; Johannes G. Wittig; Alireza Ghamari; Manami Maeda; Tamara A. Dailey; Hector A. Bergonia; Martin D. Kafina; Emma E. Coughlin; Catherine E. Minogue; Alexander S. Hebert; Liangtao Li; Jerry Kaplan; Harvey F. Lodish; Daniel E. Bauer; Stuart H. Orkin; Alan Cantor; Takahiro Maeda; John D. Phillips; Joshua J. Coon; David J. Pagliarini; Harry A. Dailey; Barry H. Paw

Heme is required for survival of all cells, and in most eukaryotes, is produced through a series of eight enzymatic reactions. Although heme production is critical for many cellular processes, how it is coupled to cellular differentiation is unknown. Here, using zebrafish, murine, and human models, we show that erythropoietin (EPO) signaling, together with the GATA1 transcriptional target, AKAP10, regulates heme biosynthesis during erythropoiesis at the outer mitochondrial membrane. This integrated pathway culminates with the direct phosphorylation of the crucial heme biosynthetic enzyme, ferrochelatase (FECH) by protein kinase A (PKA). Biochemical, pharmacological, and genetic inhibition of this signaling pathway result in a block in hemoglobin production and concomitant intracellular accumulation of protoporphyrin intermediates. Broadly, our results implicate aberrant PKA signaling in the pathogenesis of hematologic diseases. We propose a unifying model in which the erythroid transcriptional program works in concert with post-translational mechanisms to regulate heme metabolism during normal development. DOI: http://dx.doi.org/10.7554/eLife.24767.001


Science | 2007

Regulation of B Versus T Lymphoid Lineage Fate Decision by the Proto-Oncogene LRF

Takahiro Maeda; Taha Merghoub; Robin M. Hobbs; Lin Dong; Manami Maeda; Johannes L. Zakrzewski; Marcel R.M. van den Brink; Arthur Zelent; Hirokazu Shigematsu; Koichi Akashi; Julie Teruya-Feldstein; Giorgio Cattoretti; Pier Paolo Pandolfi


Blood | 2008

LRF Regulates Self-Renewal of Hematopoietic Stem Cells by Blocking Notch1-Mediated T Cell Differentiation

Sung-Uk Lee; Manami Maeda; Nagisa Sakurai; Julie Teruya-Feldstein; Freddy Radtke; Takahiro Maeda


Cancer Cell | 2018

Genome-wide CRISPR-Cas9 Screen Identifies Leukemia-Specific Dependence on a Pre-mRNA Metabolic Pathway Regulated by DCPS

Takuji Yamauchi; Takeshi Masuda; Matthew C. Canver; Michael Seiler; Yuichiro Semba; Mohammad Shboul; Mohammed Al-Raqad; Manami Maeda; Vivien A.C. Schoonenberg; Mitchel Alfonza Cole; Claudio Macias-Trevino; Yuichi Ishikawa; Qiuming Yao; Michitaka Nakano; Fumio Arai; Stuart H. Orkin; Bruno Reversade; Silvia Buonamici; Luca Pinello; Koichi Akashi; Daniel E. Bauer; Takahiro Maeda

Collaboration


Dive into the Manami Maeda's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sung-Uk Lee

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Julie Teruya-Feldstein

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Nagisa Sakurai

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Min Li

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Freddy Radtke

École Polytechnique Fédérale de Lausanne

View shared research outputs
Researchain Logo
Decentralizing Knowledge