Network


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

Hotspot


Dive into the research topics where Hiroko Nakatsukasa is active.

Publication


Featured researches published by Hiroko Nakatsukasa.


Nature | 2014

Transcription factor achaete-scute homologue 2 initiates follicular T-helper-cell development

Xindong Liu; Xin Chen; Bo Zhong; Aibo Wang; Xiaohu Wang; Fuliang Chu; Roza Nurieva; Xiaowei Yan; Ping Chen; Laurens G. van der Flier; Hiroko Nakatsukasa; Sattva S. Neelapu; WanJun Chen; Hans Clevers; Qiang Tian; Hai Qi; Lai Wei; Chen Dong

In immune responses, activated T cells migrate to B-cell follicles and develop into follicular T-helper (TFH) cells, a recently identified subset of CD4+ T cells specialized in providing help to B lymphocytes in the induction of germinal centres. Although Bcl6 has been shown to be essential in TFH-cell function, it may not regulate the initial migration of T cells or the induction of the TFH program, as exemplified by C-X-C chemokine receptor type 5 (CXCR5) upregulation. Here we show that expression of achaete-scute homologue 2 (Ascl2)—a basic helix–loop–helix (bHLH) transcription factor—is selectively upregulated in TFH cells. Ectopic expression of Ascl2 upregulates CXCR5 but not Bcl6, and downregulates C-C chemokine receptor 7 (CCR7) expression in T cells in vitro, as well as accelerating T-cell migration to the follicles and TFH-cell development in vivo in mice. Genome-wide analysis indicates that Ascl2 directly regulates TFH-related genes whereas it inhibits expression of T-helper cell 1 (TH1) and TH17 signature genes. Acute deletion of Ascl2, as well as blockade of its function with the Id3 protein in CD4+ T cells, results in impaired TFH-cell development and germinal centre response. Conversely, mutation of Id3, known to cause antibody-mediated autoimmunity, greatly enhances TFH-cell generation. Thus, Ascl2 directly initiates TFH-cell development.


Science Translational Medicine | 2014

In Vivo-Generated Antigen-Specific Regulatory T Cells Treat Autoimmunity Without Compromising Antibacterial Immune Response

Shimpei Kasagi; Pin Zhang; Li Che; Brittany Abbatiello; Takashi Maruyama; Hiroko Nakatsukasa; Peter Zanvit; Wenwen Jin; Joanne E. Konkel; WanJun Chen

Antigen-specific regulatory T cells induced in vivo have therapeutic effects in mice with established autoimmune diseases. Rebuilding Immunity Sometimes for a sports team to pull out of a slump, they need to rebuild—trade the more experienced players, however good, and start over with a fresh group of youngsters. Kasagi et al. now use the same approach to restore tolerance in mice with established autoimmune disease. They induced apoptosis of immune cells in mice with either experimental autoimmune encephalomyelitis or nonobese diabetes. The authors then introduced autoantigenic peptides, which resulted in antigen-specific regulatory T cell differentiation in vivo. These cells suppressed the autoimmune response without affecting the immune response to a bacterial antigen. They found that the apoptotic cells induced phagocytes to produce transforming growth factor–β, which was critical for the induction of the antigen-specific regulatory T cells. If these data hold true in humans, this may be a champion approach for treating autoimmunity. Harnessing regulatory T (Treg) cells is a promising approach for treating autoimmune disease. However, inducing antigen-specific Treg cells that target inflammatory immune cells without compromising beneficial immune responses has remained an unmet challenge. We developed a pathway to generate autoantigen-specific Treg cells in vivo, which showed therapeutic effects on experimental autoimmune encephalomyelitis and nonobese diabetes in mice. Specifically, we induced apoptosis of immune cells by systemic sublethal irradiation or depleted B and CD8+ T cells with specific antibodies and then administered autoantigenic peptides in mice with established autoimmune diseases. We demonstrated mechanistically that apoptotic cells triggered professional phagocytes to produce transforming growth factor–β, under which the autoantigenic peptides directed naïve CD4+ T cells to differentiate into Foxp3+ Treg cells instead of into T effector cells in vivo. These antigen-specific Treg cells specifically ameliorated autoimmunity without compromising immune responses to bacterial antigen. We have thus successfully generated antigen-specific Treg cells with therapeutic activity toward autoimmunity. The findings may lead to the development of antigen-specific Treg cell–mediated immunotherapy for multiple sclerosis and type 1 diabetes and also other autoimmune diseases.


Nature Communications | 2015

Antibiotics in neonatal life increase murine susceptibility to experimental psoriasis

Peter Zanvit; Joanne E. Konkel; Xue Jiao; Shimpei Kasagi; Dunfang Zhang; Ruiqing Wu; Cheryl Chia; Nadim J. Ajami; Daniel P. Smith; Joseph F. Petrosino; Brittany Abbatiello; Hiroko Nakatsukasa; Qianming Chen; Yasmine Belkaid; Zi Jiang Chen; WanJun Chen

Psoriasis is an inflammatory skin disease affecting ∼2% of the worlds population, but the aetiology remains incompletely understood. Recently, microbiota have been shown to differentially regulate the development of autoimmune diseases, but their influence on psoriasis is incompletely understood. We show here that adult mice treated with antibiotics that target Gram-negative and Gram-positive bacteria develop ameliorated psoriasiform dermatitis induced by imiquimod, with decreased pro-inflammatory IL-17- and IL-22-producing T cells. Surprisingly, mice treated neonatally with these antibiotics develop exacerbated psoriasis induced by imiquimod or recombinant IL-23 injection when challenged as adults, with increased IL-22-producing γδ+ T cells. 16S rRNA gene compositional analysis reveals that neonatal antibiotic-treatment dysregulates gut and skin microbiota in adults, which is associated with increased susceptibility to experimental psoriasis. This link between neonatal antibiotic-mediated imbalance in microbiota and development of experimental psoriasis provides precedence for further investigation of its specific aetiology as it relates to human psoriasis.


Nature Immunology | 2015

The DNA-binding inhibitor Id3 regulates IL-9 production in CD4(+) T cells.

Hiroko Nakatsukasa; Dunfang Zhang; Takashi Maruyama; Hua Chen; Kairong Cui; Masaki Ishikawa; Lisa Deng; Peter Zanvit; Eric Tu; Wenwen Jin; Brittany Abbatiello; Nathan Goldberg; Qianming Chen; Lingyun Sun; Keji Zhao; WanJun Chen

The molecular mechanisms by which signaling via transforming growth factor-β (TGF-β) and interleukin 4 (IL-4) control the differentiation of CD4+ IL-9-producing helper T cells (TH9 cells) remain incompletely understood. We found here that the DNA-binding inhibitor Id3 regulated TH9 differentiation, as deletion of Id3 increased IL-9 production from CD4+ T cells. Mechanistically, TGF-β1 and IL-4 downregulated Id3 expression, and this process required the kinase TAK1. A reduction in Id3 expression enhanced binding of the transcription factors E2A and GATA-3 to the Il9 promoter region, which promoted Il9 transcription. Notably, Id3-mediated control of TH9 differentiation regulated anti-tumor immunity in an experimental melanoma-bearing model in vivo and also in human CD4+ T cells in vitro. Thus, our study reveals a previously unrecognized TAK1–Id3–E2A–GATA-3 pathway that regulates TH9 differentiation.


Blood | 2013

PARP-1 regulates expression of TGF-β receptors in T cells

Pin Zhang; Hiroko Nakatsukasa; Eric Tu; Shimpei Kasagi; Kairong Cui; Masaki Ishikawa; Joanne E. Konkel; Takashi Maruyama; Gang Wei; Brittany Abbatiello; Zhao-Qi Wang; Keji Zhao; WanJun Chen

Transforming growth factor-β (TGF-β) receptors (TβRs) are essential components for TGF-β signal transduction in T cells, yet the mechanisms by which the receptors are regulated remain poorly understood. We show here that Poly(ADP-ribose) polymerase-1 (PARP-1) regulates TGF-β receptor I (TβRI) and II (TβRII) expression in CD4(+) T cells and subsequently affects Smad2/3-mediated TGF-β signal transduction. Inhibition of PARP-1 led to the upregulation of both TβRI and TβRII, yet the underlying molecular mechanisms were distinct. PARP-1 selectively bound to the promoter of TβRII, whereas the enzymatic activity of PARP-1 was responsible for the inhibition of TβRI expression. Importantly, inhibition of PARP-1 also enhanced expression of TβRs in human CD4(+) T cells. Thus, PARP-1 regulates TβR expression and TGF-β signaling in T cells.


Nature Medicine | 2017

D -mannose induces regulatory T cells and suppresses immunopathology

Dunfang Zhang; Cheryl Chia; Xue Jiao; Wenwen Jin; Shimpei Kasagi; Ruiqing Wu; Joanne E. Konkel; Hiroko Nakatsukasa; Peter Zanvit; Nathan Goldberg; Qianming Chen; Lingyun Sun; Zi Jiang Chen; WanJun Chen

D-mannose, a C-2 epimer of glucose, exists naturally in many plants and fruits, and is found in human blood at concentrations less than one-fiftieth of that of glucose. However, although the roles of glucose in T cell metabolism, diabetes and obesity are well characterized, the function of D-mannose in T cell immune responses remains unknown. Here we show that supraphysiological levels of D-mannose safely achievable by drinking-water supplementation suppressed immunopathology in mouse models of autoimmune diabetes and airway inflammation, and increased the proportion of Foxp3+ regulatory T cells (Treg cells) in mice. In vitro, D-mannose stimulated Treg cell differentiation in human and mouse cells by promoting TGF-β activation, which in turn was mediated by upregulation of integrin αvβ8 and reactive oxygen species generated by increased fatty acid oxidation. This previously unrecognized immunoregulatory function of D-mannose may have clinical applications for immunopathology.


Nature Communications | 2017

Notch-mediated conversion of activated T cells into stem cell memory-like T cells for adoptive immunotherapy

Taisuke Kondo; Rimpei Morita; Yuumi Okuzono; Hiroko Nakatsukasa; Takashi Sekiya; Shunsuke Chikuma; Takashi Shichita; Mitsuhiro Kanamori; Masato Kubo; Keiko Koga; Takahiro Miyazaki; Yoshiaki Kassai; Akihiko Yoshimura

Adoptive T-cell immunotherapy is a promising approach to cancer therapy. Stem cell memory T (TSCM) cells have been proposed as a class of long-lived and highly proliferative memory T cells. CD8+ TSCM cells can be generated in vitro from naive CD8+ T cells via Wnt signalling; however, methods do not yet exist for inducing TSCM cells from activated or memory T cells. Here, we show a strategy for generating TSCM-like cells in vitro (iTSCM cells) from activated CD4+ and CD8+ T cells in mice and humans by coculturing with stromal cells that express a Notch ligand. iTSCM cells lose PD-1 and CTLA-4 expression, and produce a large number of tumour-specific effector cells after restimulation. This method could therefore be used to generate antigen-specific effector T cells for adoptive immunotherapy.


Scientific Reports | 2018

IL-6, IL-17 and Stat3 are required for auto-inflammatory syndrome development in mouse

Takatsugu Oike; Hiroya Kanagawa; Yuiko Sato; Tami Kobayashi; Hiroko Nakatsukasa; Kana Miyamoto; Satoshi Nakamura; Yosuke Kaneko; Shu Kobayashi; Kengo Harato; Akihiko Yoshimura; Yoichiro Iwakura; Tsutomu Takeuchi; Morio Matsumoto; Masaya Nakamura; Yasuo Niki; Takeshi Miyamoto

Auto-inflammatory syndrome, a condition clinically distinct from rheumatoid arthritis, is characterized by systemic inflammation in tissues such as major joints, skin, and internal organs. Autonomous innate-immune activation is thought to promote this inflammation, but underlying pathological mechanisms have not been clarified nor are treatment strategies established. Here, we newly established a mouse model in which IL-1 signaling is conditionally activated in adult mice (hIL-1 cTg) and observed phenotypes similar to those seen in auto-inflammatory syndrome patients. In serum of hIL-1 cTg mice, IL-6 and IL-17 levels significantly increased, and signal transducer and activator of transcription 3 (Stat3) was activated in joints. When we crossed hIL-1 cTg with either IL-6- or IL-17-deficient mice or with Stat3 conditional knockout mice, phenotypes seen in hIL-1 cTg mice were significantly ameliorated. Thus, IL-6, IL-17 and Stat3 all represent potential therapeutic targets for this syndrome.


eLS | 2015

TGF‐β in the Immune Response

Hiroko Nakatsukasa; Eric Tu; Cheryl Chia; WanJun Chen


Archive | 2013

receptors in T cells β PARP-1 regulates expression of TGF-

Takashi Maruyama; Brittany Abbatiello; Zhao-Qi Wang; Keji Zhao; Hiroko Nakatsukasa; Eric Tu; Shimpei Kasagi; Kairong Cui; Masaki Ishikawa; E Joanne

Collaboration


Dive into the Hiroko Nakatsukasa's collaboration.

Top Co-Authors

Avatar

WanJun Chen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Brittany Abbatiello

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Takashi Maruyama

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Eric Tu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Joanne E. Konkel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Peter Zanvit

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cheryl Chia

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dunfang Zhang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kairong Cui

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge