Network


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

Hotspot


Dive into the research topics where Taku Naito is active.

Publication


Featured researches published by Taku Naito.


Molecular Cell | 2002

The CD8α Gene Locus Is Regulated by the Ikaros Family of Proteins

Nicola Harker; Taku Naito; Marta Cortes; Arnd Hostert; Sandra Hirschberg; Mauro Tolaini; Kathleen Roderick; Katia Georgopoulos; Dimitris Kioussis

Abstract Ikaros family members are important regulatory factors in lymphocyte development. Here we show that Ikaros may play an important role in CD4 versus CD8 lineage commitment decisions by demonstrating: (1) that it binds to regulatory elements in the endogenous CD8α locus in vivo using thymocyte chromatin immunoprecipitations, (2) that Ikaros suppresses position effect variegation of transgenes driven by CD8 regulatory elements, and (3) that mice with reduced levels of Ikaros and Aiolos show an apparent increase in CD4 populations with immature phenotype, i.e., cells that failed to activate the CD8α gene locus. We propose that Ikaros family members function as activators of the CD8α gene locus and that their associated activities are critical for appropriate chromatin remodeling transitions during thymocyte differentiation and lineage commitment.


Genes & Development | 2008

The role of the chromatin remodeler Mi-2β in hematopoietic stem cell self-renewal and multilineage differentiation

Toshimi Yoshida; Idit Hazan; Jiangwen Zhang; Samuel Y. Ng; Taku Naito; Hugo J. Snippert; Elizabeth J. Heller; Xiaoqing Qi; Lee N. Lawton; Christine J. Williams; Katia Georgopoulos

The ability of somatic stem cells to self-renew and differentiate into downstream lineages is dependent on specialized chromatin environments that keep stem cell-specific genes active and key differentiation factors repressed but poised for activation. The epigenetic factors that provide this type of regulation remain ill-defined. Here we provide the first evidence that the SNF2-like ATPase Mi-2beta of the Nucleosome Remodeling Deacetylase (NuRD) complex is required for maintenance of and multilineage differentiation in the early hematopoietic hierarchy. Shortly after conditional inactivation of Mi-2beta, there is an increase in cycling and a decrease in quiescence in an HSC (hematopoietic stem cell)-enriched bone marrow population. These cycling mutant cells readily differentiate into the erythroid lineage but not into the myeloid and lymphoid lineages. Together, these effects result in an initial expansion of mutant HSC and erythroid progenitors that are later depleted as more differentiated proerythroblasts accumulate at hematopoietic sites exhibiting features of erythroid leukemia. Examination of gene expression in the mutant HSC reveals changes in the expression of genes associated with self-renewal and lineage priming and a pivotal role of Mi-2beta in their regulation. Thus, Mi-2beta provides the hematopoietic system with immune cell capabilities as well as with an extensive regenerative capacity.


Nature Immunology | 2012

Harnessing of the nucleosome-remodeling-deacetylase complex controls lymphocyte development and prevents leukemogenesis

Jiangwen Zhang; Audrey F. Jackson; Taku Naito; Marei Dose; John Seavitt; Feifei Liu; Elizabeth J. Heller; Mariko Kashiwagi; Toshimi Yoshida; Fotini Gounari; Howard T. Petrie; Katia Georgopoulos

Cell fate depends on the interplay between chromatin regulators and transcription factors. Here we show that activity of the Mi-2β nucleosome-remodeling and histone-deacetylase (NuRD) complex was controlled by the Ikaros family of lymphoid lineage–determining proteins. Ikaros, an integral component of the NuRD complex in lymphocytes, tethered this complex to active genes encoding molecules involved in lymphoid differentiation. Loss of Ikaros DNA-binding activity caused a local increase in chromatin remodeling and histone deacetylation and suppression of lymphoid cell–specific gene expression. Without Ikaros, the NuRD complex also redistributed to transcriptionally poised genes that were not targets of Ikaros (encoding molecules involved in proliferation and metabolism), which induced their reactivation. Thus, release of NuRD from Ikaros regulation blocks lymphocyte maturation and mediates progression to a leukemic state by engaging functionally opposing epigenetic and genetic networks.


International Immunology | 2011

Transcriptional control of T-cell development

Taku Naito; Hirokazu Tanaka; Yoshinori Naoe; Ichiro Taniuchi

T lymphocytes, which are central players in orchestrating immune responses, consist of several subtypes with distinct functions. The thymus is an organ where hematopoietic progenitors undergo sequential developmental processes to give rise to this variety of T-cell subsets with diverse antigen specificity. In the periphery, naive T cells further differentiate into effector cells upon encountering antigens. There are several developmental checkpoints during T-cell development, where regulation by a combination of transcription factors imprints specific functional properties on precursors. The transcription factors E2A, GATA-binding protein 3 (Gata3) and RUNT-related transcription factor (Runx) are involved at various stages in the differentiation of double-negative thymocytes and in β-selection, as are transcription factors from the Notch signaling pathway; other transcription factors such as B-cell lymphoma/leukemia 11b (Bcl11b), myeloblastosis viral oncogene homolog (Myb) and inhibitor of DNA binding 3 (Id3) are involved at specific stages. Differentiation of T cells into helper versus cytotoxic cells involves not only antagonistic interplay between Runx and T(h) inducing POZ-Kruppel factor (ThPOK) but also complex interactions between MAZR, Gata3 and Myb in the activation and silencing of genes such as Cd4 and Cd8 as well as the gene that encodes ThPOK itself. A wide range of well-defined transcription factors, including signal transducer and activator of transcriptions (STATs), T-bet, Gata3, nuclear factor of activated T cell (NFAT), adaptor-related protein complex 1 (AP-1) and nuclear factor κB (NF-κB), are known to shape T(h)1/T(h)2 differentiation. Runx and Gata3 also operate in this process, as do c-Maf and recombining binding protein for immunoglobulin Jκ region (RBP-J) and the chromatin-reorganizing protein special AT-rich sequence-binding protein 1 (SATB1). In this review, we briefly discuss how T-cell characteristics are acquired and become divergent from the point of view of transcriptional regulation.


Blood | 2013

Transcriptional regulation of the Ikzf1 locus.

Toshimi Yoshida; Esther Landhuis; Marei Dose; Idit Hazan; Jiangwen Zhang; Taku Naito; Audrey F. Jackson; Jeffrey H. Wu; Elizabeth A. Perotti; Christoph Kaufmann; Fotini Gounari; Bruce A. Morgan; Katia Georgopoulos

Ikaros is a critical regulator of lymphocyte development and homeostasis; thus, understanding its transcriptional regulation is important from both developmental and clinical perspectives. Using a mouse transgenic reporter approach, we functionally characterized a network of highly conserved cis-acting elements at the Ikzf1 locus. We attribute B-cell and myeloid but not T-cell specificity to the main Ikzf1 promoter. Although this promoter was unable to counter local chromatin silencing effects, each of the 6 highly conserved Ikzf1 intronic enhancers alleviated silencing. Working together, the Ikzf1 enhancers provided locus control region activity, allowing reporter expression in a position and copy-independent manner. Only 1 of the Ikzf1 enhancers was responsible for the progressive upregulation of Ikaros expression from hematopoietic stem cells to lymphoid-primed multipotent progenitors to T-cell precursors, which are stages of differentiation dependent on Ikaros for normal outcome. Thus, Ikzf1 is regulated by both epigenetic and transcriptional factors that target its enhancers in both redundant and specific fashions to provide an expression profile supportive of normal lymphoid lineage progression and homeostasis. Mutations in the Ikzf1 regulatory elements and their interacting factors are likely to have adverse effects on lymphopoiesis and contribute to leukemogenesis.


International Immunology | 2010

The network of transcription factors that underlie the CD4 versus CD8 lineage decision

Taku Naito; Ichiro Taniuchi

Virtually all mature T cells are CD4(+)CD8(-) or CD4(-)CD8(+) and this not only is their most important surface-phenotype distinction but also has crucial functional consequences for the entire immune response. Both subsets arise from double-positive thymocytes, and much has been learned about the molecular events that govern this lineage bifurcation process. As detailed in this review, the signaling pathways and specific molecules that control this process are now being discovered. In particular, the transcription factors ThPOK (T-helper inducing POZ-Kruppel factor) and Runx3 have emerged as the crucial regulators of helper lineage commitment and the cytotoxic lineage, respectively. This article describes their antagonistic interaction that is an important mechanism of the lineage specification, as well as the hierarchy and importance of several other transcription factors and cytokine signals in the network of pathways that govern thymocyte helper/cytotoxic lineage commitment.


Journal of Immunology | 2015

SATB1 Plays a Critical Role in Establishment of Immune Tolerance

Motonari Kondo; Yuriko Tanaka; Taku Kuwabara; Taku Naito; Terumi Kohwi-Shigematsu; Akiko Watanabe

Special AT-rich sequence binding protein 1 (SATB1) is a genome organizer that is expressed by T cells. T cell development is severely impaired in SATB1 null mice; however, because SATB1 null mice die by 3 wk of age, the roles of SATB1 in T cell development have not been well clarified. In this study, we generated and analyzed SATB1 conditional knockout (cKO) mice, in which the SATB1 gene was deleted from all hematopoietic cells. T cell numbers were reduced in these mice, mainly because of a deficiency in positive selection at the CD4+CD8+ double-positive stage during T cell development in the thymus. We also found that SATB1 cKO mice developed autoimmune diseases within 16 wk after birth. In SATB1 cKO mice, the numbers of Foxp3+ regulatory T (Treg) cells were significantly reduced at 2 wk of age compared with wild-type littermates. Although the numbers gradually increased upon aging, Treg cells in SATB1 cKO mice were still less than those in wild-type littermates at adulthood. Suppressive functions of Treg cells, which play a major role in establishment of peripheral tolerance, were also affected in the absence of SATB1. In addition, negative selection during T cell development in the thymus was severely impaired in SATB1 deficient mice. These results suggest that SATB1 plays an essential role in establishment of immune tolerance.


The EMBO Journal | 2013

Epigenetic Thpok silencing limits the time window to choose CD4+ helper-lineage fate in the thymus

Hirokazu Tanaka; Taku Naito; Sawako Muroi; Wooseok Seo; Risa Chihara; Chizuko Miyamoto; Ryo Kominami; Ichiro Taniuchi

CD4+ helper and CD8+ cytotoxic T cells differentiate from common precursors in the thymus after T‐cell receptor (TCR)‐mediated selection. Commitment to the helper lineage depends on persistent TCR signals and expression of the ThPOK transcription factor, whereas a ThPOK cis‐regulatory element, ThPOK silencer, represses Thpok gene expression during commitment to the cytotoxic lineage. Here, we show that silencer‐mediated alterations of chromatin structures in cytotoxic‐lineage thymocytes establish a repressive state that is epigenetically inherited in peripheral CD8+ T cells even after removal of the silencer. When silencer activity is enhanced in helper‐lineage cells, by increasing its copy number, a similar heritable Thpok silencing occurs. Epigenetic locking of the Thpok locus may therefore be an independent event from commitment to the cytotoxic lineage. These findings imply that long‐lasting TCR signals are needed to establish stable Thpok expression activity to commit to helper T‐cell fate and that full commitment to the helper lineage requires persistent reversal of silencer activity during a particular time window.


Journal of Immunology | 2011

Pre-TCR Signaling and CD8 Gene Bivalent Chromatin Resolution during Thymocyte Development

Nicola Harker; Anna Garefalaki; Ursula Menzel; Eleni Ktistaki; Taku Naito; Katia Georgopoulos; Dimitris Kioussis

The CD8 gene is silent in CD4−CD8− double-negative thymocytes, expressed in CD4+CD8+ double-positive cells, and silenced in cells committing to the CD4+ single-positive (SP) lineage, remaining active in the CD8+ SP lineage. In this study, we show that the chromatin of the CD8 locus is remodeled in C57BL/6 and B6/J Rag1−/− MOM double-negative thymocytes as indicated by DNaseI hypersensitivity and widespread bivalent chromatin marks. Pre-TCR signaling coincides with chromatin bivalency resolution into monovalent activating modifications in double-positive and CD8 SP cells. Shortly after commitment to CD4 SP cell lineage, monovalent repressive characteristics and chromatin inaccessibility are established. Differential binding of Ikaros, NuRD, and heterochromatin protein 1α on the locus during these processes may participate in the complex regulation of CD8.


Nature Communications | 2014

Histone acetylation mediated by Brd1 is crucial for Cd8 gene activation during early thymocyte development

Yuta Mishima; Changshan Wang; Satoru Miyagi; Atsunori Saraya; Hiroyuki Hosokawa; Makiko Mochizuki-Kashio; Yaeko Nakajima-Takagi; Shuhei Koide; Masamitsu Negishi; Goro Sashida; Taku Naito; Tomoyuki Ishikura; Atsushi Onodera; Toshinori Nakayama; Daniel G. Tenen; Naoto Yamaguchi; Haruhiko Koseki; Ichiro Taniuchi; Atsushi Iwama

During T-cell development, Cd8 expression is controlled via dynamic regulation of its cis-regulatory enhancer elements. Insufficiency of enhancer activity causes variegated Cd8 expression in CD4(+)CD8(+) double-positive (DP) thymocytes. Brd1 is a subunit of the Hbo1 histone acetyltransferase (HAT) complex responsible for acetylation of histone H3 at lysine 14 (H3K14). Here we show that deletion of Brd1 in haematopoietic progenitors causes variegated expression of Cd8, resulting in the appearance of CD4(+)CD8(-)TCRβ(-/low) thymocytes indistinguishable from DP thymocytes in their properties. Biochemical analysis confirms that Brd1 forms a HAT complex with Hbo1 in thymocytes. ChIP analysis demonstrates that Brd1 localizes at the known enhancers in the Cd8 genes and is responsible for acetylation at H3K14. These findings indicate that the Brd1-mediated HAT activity is crucial for efficient activation of Cd8 expression via acetylation at H3K14, which serves as an epigenetic mark that promotes the recruitment of transcription machinery to the Cd8 enhancers.

Collaboration


Dive into the Taku Naito'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

Pablo Gómez-del Arco

Centro Nacional de Investigaciones Cardiovasculares

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge