研究者業績

平原 潔

ヒラハラ キヨシ  (Kiyoshi Hirahara)

基本情報

所属
千葉大学 大学院医学研究院先端研究部門高次機能治療学研究講座免疫発生学 教授
学位
博士(医学)(新潟大学)

研究者番号
00707193
J-GLOBAL ID
201801001136657946
researchmap会員ID
B000302077

2001年新潟大学医学部卒。2008年同大学院医歯学総合研究科修了。医学博士。
2009年米国国立衛生研究所 Visiting Fellow、2013年千葉大学大学院医学研究院特任准教授を経て、2016年より千葉大学大学院医学研究院准教授、2022年より同教授(現職)。
内科医(特に呼吸器内科医)として、5年以上臨床医療に携わった後、基礎医学研究の世界へ飛び込みました。ベッドサイドへのフィ-ドバックを目標とした基礎研究確立を希い、日々研究活動に勤しんでいます。

専門分野:免疫学、呼吸器内科学
現在の主要なテーマ:
*肺線維化における獲得免疫系の役割
*組織常在性記憶CD4 T細胞の分化・維持機構
*アレルギー疾患の病態形成機構
*肺における免疫応答一般


論文

 92
  • Shigeru Iwata, Yohei Mikami, Hong-Wei Sun, Stephen R. Brooks, Dragana Jankovic, Kiyoshi Hirahara, Atsushi Onodera, Han-Yu Shih, Takeshi Kawabe, Kan Jiang, Toshinori Nakayama, Alan Sher, John J. O'Shea, Fred P. Davis, Yuka Kanno
    IMMUNITY 46(6) 983-+ 2017年6月  査読有り
    Host defense requires the specification of CD4(+) helper T (Th) cells into distinct fates, including Th1 cells that preferentially produce interferon-gamma (IFN-gamma). IFN-gamma, a member of a large family of antipathogenic and anti-tumor IFNs, induces T-bet, a lineage-defining transcription factor for Th1 cells, which in turn supports IFN-gamma production in a feed-forward manner. Herein, we show that a cellintrinsic role of T-bet influences how T cells perceive their secreted product in the environment. In the absence of T-bet, IFN-gamma aberrantly induced a type I IFN transcriptomic program. T-bet preferentially repressed genes and pathways ordinarily activated by type I IFNs to ensure that its transcriptional response did not evoke an aberrant amplification of type I IFN signaling circuitry, otherwise triggered by its own product. Thus, in addition to promoting Th1 effector commitment, T-bet acts as a repressor in differentiated Th1 cells to prevent abberant autocrine type I IFN and downstream signaling.
  • Juergen Brueck, Julia Holstein, Ivana Glocova, Ursula Seidel, Julia Geisel, Toshio Kanno, Jin Kumagai, Naoko Mato, Stephan Sudowe, Katja Widmaier, Tobias Sinnberg, Amir S. Yazdi, Franziska C. Eberle, Kiyoshi Hirahara, Toshinori Nakayama, Martin Roecken, Kamran Ghoreschi
    SCIENTIFIC REPORTS 7 44482 2017年3月  査読有り
    The nutritional curcumin (CUR) is beneficial in cell-mediated autoimmune diseases. The molecular mechanisms underlying this food-mediated silencing of inflammatory immune responses are poorly understood. By investigating antigen-specific immune responses we found that dietary CUR impairs the differentiation of Th1/Th17 cells in vivo during encephalomyelitis and instead promoted Th2 cells. In contrast, feeding CUR had no inhibitory effect on ovalbumin-induced airway inflammation. Mechanistically, we found that CUR induces an anti-inflammatory phenotype in dendritic cells (DC) with enhanced STAT3 phosphorylation and suppressed expression of Il12b and Il23a. On the molecular level CUR readily induced NRF2-sensitive heme oxygenase 1 (HO-1) mRNA and protein in LPS-activated DC. HO-1 enhanced STAT3 phosphorylation, which enriched to Il12b and Il23a loci and negatively regulated their transcription. These findings demonstrate the underlying mechanism through which a nutritional can interfere with the immune response. CUR silences IL-23/Th17-mediated pathology by enhancing HO-1/STAT3 interaction in DC.
  • Toshinori Nakayama, Kiyoshi Hirahara, Atsushi Onodera, Yusuke Endo, Hiroyuki Hosokawa, Kenta Shinoda, Damon J. Tumes, Yoshitaka Okamoto
    ANNUAL REVIEW OF IMMUNOLOGY, VOL 35 35 53-84 2017年  査読有り
    Helper T (Th) cell subsets direct immune responses by producing signature cytokines. Th2 cells produce IL-4, IL-5, and IL-13, which are important in humoral immunity and protection from helminth infection and are central to the pathogenesis of many allergic inflammatory diseases. Molecular analysis of Th2 cell differentiation and maintenance of function has led to recent discoveries that have refined our understanding of Th2 cell biology. Epigenetic regulation of Gata3 expression by chromatin remodeling complexes such as Polycomb and Trithorax is crucial for maintaining Th2 cell identity. In the context of allergic diseases, memory-type pathogenic Th2 cells have been identified in both mice and humans. To better understand these disease-driving cell populations, we have developed a model called the pathogenic Th population disease induction model. The concept of defined subsets of pathogenic Th cells may spur new, effective strategies for treating intractable chronic inflammatory disorders.
  • Kiyoshi Hirahara, Daniella Schwartz, Massimo Gadina, Yuka Kanno, John J. O'Shea
    CURRENT OPINION IN IMMUNOLOGY 43 89-97 2016年12月  査読有り
    Cytokines represent structurally diverse soluble factors with critical roles in normal immune function and the pathogenesis of autoimmunity. The emergence of many successful biological therapies targeting cytokines and cytokine receptors exemplifies the importance of cytokines in driving human autoimmune disease; unsurprisingly, there is no paucity of reviews on this subject. Nonetheless, many patients with autoimmune disease do not respond to biologicals, and cure remains an unmet goal. Thus, targeting the intracellular pathways employed by cytokines provides new therapeutic opportunities. A subset of cytokines utilizes the Janus kinase-signal transducer of activators of transcription (JAK-STAT) pathway as a mode of signal transduction. First generation JAK inhibitors (jakinibs) are used to treat rheumatologic disease, and second-generation jakinibs are being developed. Simultaneously, rapid advances are being made in our understanding of the genomic and epigenomic impact of cytokines. In this review, we will briefly review the role of JAKSTAT-dependent cytokines in immune-mediated disease, the current status of Jakinibs, and future possibilities for therapeutic intervention using genomic insights.
  • Toshinori Nakayama, Yusuke Endo, Kenta Shinoda, Kiyoshi Hirahara
    CORNEA 35 S8-S8 2016年11月  査読有り
  • Koji Hayashizaki, Motoko Y. Kimura, Koji Tokoyoda, Hiroyuki Hosokawa, Kenta Shinoda, Kiyoshi Hirahara, Tomomi Ichikawa, Atsushi Onodera, Asami Hanazawa, Chiaki Iwamura, Jungo Kakuta, Kenzo Muramoto, Shinichiro Motohashi, Damon J. Tumes, Tomohisa Iinuma, Heizaburo Yamamoto, Yuzuru Ikehara, Yoshitaka Okamoto, Toshinori Nakayama
    Science Immunology 1(3) eaaf9154-eaaf9154 2016年9月  査読有り
  • Kenta Shinoda, Kiyoshi Hirahara, Tomohisa Iinuma, Tomomi Ichikawa, Akane S. Suzuki, Kaoru Sugaya, Damon J. Tumes, Heizaburo Yamamoto, Takahiro Hara, Shizue Tani-ichie, Koichi Ikuta, Yoshitaka Okamoto, Toshinori Nakayama
    PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 113(20) E2842-E2851 2016年5月  査読有り
    Memory CD4(+) T helper (Th) cells are central to long-term protection against pathogens, but they can also be pathogenic and drive chronic inflammatory disorders. How these pathogenic memory Th cells are maintained, particularly at sites of local inflammation, remains unclear. We found that ectopic lymphoid-like structures called inducible bronchus-associated lymphoid tissue (iBALT) are formed during chronic allergic inflammation in the lung, and that memory-type pathogenic Th2 (Tpath2) cells capable of driving allergic inflammation are maintained within the iBALT structures. The maintenance of memory Th2 cells within iBALT is supported by Thy1(+) IL-7-producing lymphatic endothelial cells (LECs). The Thy1(+) IL-7-producing LECs express IL-33 and T-cell-attracting chemokines CCL21 and CCL19. Moreover, ectopic lymphoid structures consisting of memory CD4(+) T cells and IL-7(+) IL-33(+) LECs were found in nasal polyps of patients with eosinophilic chronic rhinosinusitis. Thus, Thy1(+) IL-7-producing LECs control chronic allergic airway inflammation by providing a survival niche for memory-type Tpath2 cells.
  • Franziska C. Eberle, Jürgen Brück, Julia Holstein, Kiyoshi Hirahara, Kamran Ghoreschi
    F1000Research 5 770-770 2016年4月28日  査読有り
    <p>T helper (Th) cells producing interleukin (IL)-17, IL-22, and tumor necrosis factor (TNF) form the key T cell population driving psoriasis pathogenesis. They orchestrate the inflammation in the skin that results in the proliferation of keratinocytes and endothelial cells. Besides Th17 cells, other immune cells that are capable of producing IL-17-associated cytokines participate in psoriatic inflammation. Recent advances in psoriasis research improved our understanding of the cellular and molecular players that are involved in Th17 pathology and inflammatory pathways in the skin. The inflammation-driving actions of TNF in psoriasis are already well known and antibodies against TNF are successful in the treatment of Th17-mediated psoriatic skin inflammation. A further key cytokine with potent IL-17-/IL-22-promoting properties is IL-23. Therapeutics directly neutralizing IL-23 or IL-17 itself are now extending the therapeutic spectrum of antipsoriatic agents and further developments are on the way. The enormous progress in psoriasis research allows us to control this Th17-mediated inflammatory skin disease in many patients.</p>
  • Michael Bonelli, Lisa Goeschl, Stephan Blueml, Thomas Karonitsch, Kiyoshi Hirahara, Elisabeth Ferner, Carl-Walter Steiner, Guenter Steiner, Josef S. Smolen, Clemens Scheinecker
    RHEUMATOLOGY 55(4) 710-720 2016年4月  査読有り
    Methods. Phenotypic and functional analyses of CD4(+) T cells, including CD4(+) FoxP3(+) CD25(+) regulatory T cells (Treg), from RA patients were performed before and during CTLA-4Ig therapy. In addition T cells from healthy volunteers were analysed on in vitro culture with CTLA-4Ig or anti-CD80 and anti-CD86 antibodies. Apoptotic DNA fragmentation in CD4(+) and CD4(+) FoxP3(+) T cells was measured by TUNEL staining. Results. We observed an increase in T cells, including Treg cells, after initiation of CTLA-4Ig therapy, which was linked to a downregulation of activation-associated marker molecules and CD95 on CD4(+) T cells and Treg cells. CTLA-4Ig decreased CD95-mediated cell death in vitro in a dose-dependent manner. Functional analysis of isolated Treg cells from RA patients further revealed a diminished suppression of responder T cell proliferation. This was found to be due to CTLA-4Ig-mediated blocking of CD80 and CD86 on responder T cells that led to a diminished susceptibility for Treg cell suppression. Conclusion. CTLA-4Ig therapy in RA patients exerts effects beyond the suppression of T cell activation, which has to be taken into account as an additional mechanism of CTLA-4Ig treatment.
  • Kiyoshi Hirahara, Toshinori Nakayama
    INTERNATIONAL IMMUNOLOGY 28(4) 163-171 2016年4月  査読有り
    CD4(+) T cells are crucial for directing appropriate immune responses during host defense and for the pathogenesis of inflammatory diseases. In addition to the classical biphasic model of differentiation of T-helper 1 (T(h)1) and T(h)2 cells, unexpected increases in the numbers of CD4(+) T-cell subsets, including T(h)17, T(h)9, T follicular-helper (T-fh) and T-regulatory (T-reg) cells, have been recognized. In the present review, we focus on how these various T-helper cell subsets contribute to the pathogenesis of immune-mediated inflammatory diseases. In particular, we focus on multiple sclerosis, psoriasis and asthma as typical model diseases in which multiple T-helper cell subsets have recently been suggested to play a role. We will also discuss various unique sub-populations of T-helper cells that have been identified. First, we will introduce the heterogeneous T-helper cell subsets, which are classified by their simultaneous expression of multiple key transcription factors. We will also introduce different kinds of memory-type T(h)2 cells, which are involved in the pathogenesis of chronic type-2 immune-related diseases. Finally, we will discuss the molecular mechanisms underlying the generation of the plasticity and heterogeneity of T-helper cell subsets. The latest progress in the study of T-helper cell subsets has forced us to reconsider the etiology of immune-mediated inflammatory diseases beyond the model based on the T(h)1/T(h)2 balance. To this end, we propose another model-the pathogenic T-helper population disease-induction model-as a possible mechanism for the induction and/or persistence of immune-mediated inflammatory diseases.
  • Renu Jain, Yi Chen, Yuka Kanno, Barbara Joyce-Shaikh, Golnaz Vahedi, Kiyoshi Hirahara, Wendy M. Blumenschein, Selvakumar Sukumar, Christopher J. Haines, Svetlana Sadekova, Terrill K. McClanahan, Mandy J. McGeachy, John J. O'Shea, Daniel J. Cua
    IMMUNITY 44(1) 131-142 2016年1月  査読有り
    Interleukin-23 (IL-23) is a pro-inflammatory cytokine required for the pathogenicity of T helper 17 (Th17) cells but the molecular mechanisms governing this process remain unclear. We identified the transcription factor Blimp-1 (Prdm1) as a key IL-23-induced factor that drove the inflammatory function of Th17 cells. In contrast to thymic deletion of Blimp-1, which causes T cell development defects and spontaneous autoimmunity, peripheral deletion of this transcription factor resulted in reduced Th17 activation and reduced severity of autoimmune encephalomyelitis. Furthermore, genome-wide occupancy and overexpression studies in Th17 cells revealed that Blimp-1 co-localized with transcription factors ROR gamma t, STAT-3, and p300 at the Il23r, Il17a/f, and Csf2 cytokine loci to enhance their expression. Blimp-1 also directly bound to and repressed cytokine loci Il2 and Bcl6. Taken together, our results demonstrate that Blimp-1 is an essential transcription factor downstream of IL-23 that acts in concert with RORgt to activate the Th17 inflammatory program.
  • Atsushi Onodera, Damon J. Tumes, Yukiko Watanabe, Kiyoshi Hirahara, Atsushi Kaneda, Fumihiro Sugiyama, Yutaka Suzuki, Toshinori Nakayama
    MOLECULAR AND CELLULAR BIOLOGY 35(22) 3841-3853 2015年11月  査読有り
    Trithorax group (TrxG) and Polycomb group (PcG) proteins are two mutually antagonistic chromatin modifying complexes, however, how they together mediate transcriptional counter-regulation remains unknown. Genome-wide analysis revealed that binding of Ezh2 and menin, central members of the PcG and TrxG complexes, respectively, were reciprocally correlated. Moreover, we identified a developmental change in the positioning of Ezh2 and menin in differentiated T lymphocytes compared to embryonic stem cells. Ezh2-binding upstream and menin-binding downstream of the transcription start site was frequently found at genes with higher transcriptional levels, and Ezh2-binding downstream and menin-binding upstream was found at genes with lower expression in T lymphocytes. Interestingly, of the Ezh2 and menin cooccupied genes, those exhibiting occupancy at the same position displayed greatly enhanced sensitivity to loss of Ezh2. Finally, we also found that different combinations of Ezh2 and menin occupancy were associated with expression of specific functional gene groups important for T cell development. Therefore, spatial cooperative gene regulation by the PcG and TrxG complexes may represent a novel mechanism regulating the transcriptional identity of differentiated cells.
  • Yusuke Endo, Hikari K. Asou, Nao Matsugae, Kiyoshi Hirahara, Kenta Shinoda, Damon J. Tumes, Hirotake Tokuyama, Koutaro Yokote, Toshinori Nakayama
    CELL REPORTS 12(6) 1042-1055 2015年8月  査読有り
    Chronic inflammation due to obesity contributes to the development of metabolic diseases, autoimmune diseases, and cancer. Reciprocal interactions between metabolic systems and immune cells have pivotal roles in the pathogenesis of obesity-associated diseases, although the mechanisms regulating obesity-associated inflammatory diseases are still unclear. In the present study, we performed transcriptional profiling of memory phenotype CD4 T cells in high-fat-fed mice and identified acetylCoA carboxylase 1 (ACC1, the gene product of Acaca) as an essential regulator of Th17 cell differentiation in vitro and of the pathogenicity of Th17 cells in vivo. ACC1 modulates the DNA binding of ROR gamma t to target genes in differentiating Th17 cells. In addition, we found a strong correlation between IL-17A-producing CD45RO(+)CD4 T cells and the expression of ACACA in obese subjects. Thus, ACC1 confers the appropriate function of ROR gamma t through fatty acid synthesis and regulates the obesity-related pathology of Th17 cells.
  • Xiang-Ping Yang, Kan Jiang, Kiyoshi Hirahara, Golnaz Vahedi, Behdad Afzali, Giuseppe Sciume, Michael Bonelli, Hong-Wei Sun, Dragana Jankovic, Yuka Kanno, Vittorio Sartorelli, John J. O'Shea, Arian Laurence
    SCIENTIFIC REPORTS 5 2015年6月  査読有り
    The roles of EZH2 in various subsets of CD4(+) T cells are controversial and its mechanisms of action are incompletely understood. FOXP3-positive Treg cells are a critical helper T cell subset, and dysregulation of Treg generation or function results in systemic autoimmunity. FOXP3 associates with EZH2 to mediate gene repression and suppressive function. Herein, we demonstrate that deletion of Ezh2 in CD4(+) T cells resulted in reduced numbers of Treg cells in vivo and differentiation in vitro and an increased proportion of memory CD4(+) T cells in part due to exaggerated production of effector cytokines. Furthermore, we found that both Ezh2-deficient Treg cells and T effector cells were functionally impaired in vivo: Tregs failed to constrain autoimmune colitis and T effector cells neither provided a protective response to T. gondii infection nor mediated autoimmune colitis. The dichotomous function of EZH2 in regulating differentiation and senescence in effector and regulatory T cells helps to explain the apparent existing contradictions in literature.
  • Kiyoshi Hirahara, Atsushi Onodera, Alejandro V. Villarino, Michael Bonelli, Giuseppe Sciume, Arian Laurence, Hong-Wei Sun, Stephen R. Brooks, Golnaz Vahedi, Han-Yu Shih, Gustavo Gutierrez-Cruz, Shigeru Iwata, Ryo Suzuki, Yohei Mikami, Yoshitaka Okamoto, Toshinori Nakayama, Steven M. Holland, Christopher A. Hunter, Yuka Kanno, John J. O'Shea
    IMMUNITY 42(5) 877-889 2015年5月  査読有り
    Interleukin-6 (IL-6) and IL-27 signal through a shared receptor subunit and employ the same downstream STAT transcription proteins, but yet are ascribed unique and overlapping functions. To evaluate the specificity and redundancy for these cytokines, we quantified their global transcriptomic changes and determined the relative contributions of STAT1 and STAT3 using genetic models and chromatin immunoprecipitation- sequencing (ChIP-seq) approaches. We found an extensive overlap of the transcriptomes induced by IL-6 and IL-27 and few examples in which the cytokines acted in opposition. Using STAT-deficient cells and T cells from patients with gain-of-function STAT1 mutations, we demonstrated that STAT3 is responsible for the overall transcriptional output driven by both cytokines, whereas STAT1 is the principal driver of specificity. STAT1 cannot compensate in the absence of STAT3 and, in fact, much of STAT1 binding to chromatin is STAT3 dependent. Thus, STAT1 shapes the specific cytokine signature superimposed upon STAT3's action.
  • Tomohisa Iinuma, Yoshitaka Okamoto, Heizaburo Yamamoto, Ayako Inamine-Sasaki, Yuji Ohki, Toshioki Sakurai, Urara Funakoshi, Syuji Yonekura, Daiju Sakurai, Kiyoshi Hirahara, Toshinori Nakayama
    ANNALS OF ALLERGY ASTHMA & IMMUNOLOGY 114(4) 289-298 2015年4月  査読有り
    Background: Chronic rhinosinusitis with nasal polyps (CRSwNP) is a heterogeneous disease of uncertain pathogenesis. Memory T cells acquire additional functions during the secondary response and play important roles in chronic inflammation. Objective: To investigate characteristics of tissue memory CD4(+) T cells obtained from patients with non-eosinophilic CRSwNP (NECRS) and eosinophilic CRSwNP (ECRS) by focusing on the influence of interleukin (IL)-25. Methods: Pro-allergic cytokines in tissue homogenates were measured using enzyme-linked immunosorbent assays. NP mononuclear cells and CD4(+) T cells were isolated from NPs from patients with CRSwNP. Cytokine expression and CD4(+) T-cell subpopulations were analyzed using enzyme-linked immunosorbent assay, flow cytometry, and real-time polymerase chain reaction. Results: The IL-25 level in NPs increased in patients with ECRS. IL-5 and IL-9 mRNA levels expressed by tissue CD4(+) T cells were significantly elevated in patients with ECRS. Most infiltrating CD4(+) T cells in ECRS and NECRS expressed CD45RO; however, regardless of the atopic status, high IL-17RB levels were detected in CD4(+) T cells from patients with ECRS. IL-17RB mRNA levels expressed by tissue CD4(+) T cells significantly correlated with the number of eosinophils in NPs. Elevation of IL-5 and IL-9 production was found in NP mononuclear cells from patients with ECRS, but not in those from patients with NECRS, by stimulation with IL-25 under T-cell receptor stimulation. Conclusion: Interleukin-25 and a subpopulation of tissue T-helper type 2 and 9 cells that express increased IL-17RB levels could contribute to infiltration of eosinophils in NPs and could have produced the pathologic difference between NECRS and ECRS. (C) 2015 American College of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved.
  • Yusuke Endo, Kiyoshi Hirahara, Tomohisa Iinuma, Kenta Shinoda, Damon J. Tumes, Hikari K. Asou, Nao Matsugae, Kazushige Obata-Ninomiya, Heizaburo Yamamoto, Shinichiro Motohashi, Keisuke Oboki, Susumu Nakae, Hirohisa Saito, Yoshitaka Okamoto, Toshinori Nakayama
    IMMUNITY 42(2) 294-308 2015年2月  査読有り
    Memory CD4(+) T helper (Th) cells provide long-term protection against pathogens and are essential for the development of vaccines; however, some antigen-specific memory Th cells also drive immunerelated pathology, including asthma. The mechanisms regulating the pathogenicity of memory Th cells remain poorly understood. We found that interleukin-33 (IL-33)-ST2 signals selectively licensed memory Th2 cells to induce allergic airway inflammation via production of IL-5 and that the p38 MAP kinase pathway was a central downstream target of IL-33-ST2 in memory Th2 cells. In addition, we found that IL-33 induced upregulation of IL-5 by memory CD4(+) T cells isolated from nasal polyps of patients with eosinophilic chronic rhinosinusitis. Thus, IL-33ST2-p38 signaling appears to directly instruct pathogenic memory Th2 cells to produce IL-5 and induce eosinophilic inflammation.
  • Han-Yu Shih, Giuseppe Sciume, Amanda C. Poholek, Golnaz Vahedi, Kiyoshi Hirahara, Alejandro V. Villarino, Michael Bonelli, Remy Bosselut, Yuka Kanno, Stefan A. Muljo, John J. O'Shea
    IMMUNOLOGICAL REVIEWS 261(1) 23-49 2014年9月  査読有り
    The discovery of the specification of CD4(+) helper T cells to discrete effector 'lineages' represented a watershed event in conceptualizing mechanisms of host defense and immunoregulation. However, our appreciation for the actual complexity of helper T-cell subsets continues unabated. Just as the Sami language of Scandinavia has 1000 different words for reindeer, immunologists recognize the range of fates available for a CD4(+) T cell is numerous and may be underestimated. Added to the crowded scene for helper T-cell subsets is the continuously growing family of innate lymphoid cells (ILCs), endowed with common effector responses and the previously defined 'master regulators' for CD4(+) helper T-cell subsets are also shared by ILC subsets. Within the context of this extraordinary complexity are concomitant advances in the understanding of transcriptomes and epigenomes. So what do terms like 'lineage commitment' and helper T-cell 'specification' mean in the early 21st century? How do we put all of this together in a coherent conceptual framework? It would be arrogant to assume that we have a sophisticated enough understanding to seriously answer these questions. Instead, we review the current status of the flexibility of helper T-cell responses in relation to their genetic regulatory networks and epigenetic landscapes. Recent data have provided major surprises as to what master regulators can or cannot do, how they interact with other transcription factors and impact global genome-wide changes, and how all these factors come together to influence helper cell function.
  • Yukiko Watanabe, Atsushi Onodera, Urara Kanai, Tomomi Ichikawa, Kazushige Obata-Ninomiya, Tomoko Wada, Masahiro Kiuchi, Chiaki Iwamura, Damon J. Tumes, Kenta Shinoda, Ryoji Yagi, Shinichiro Motohashi, Kiyoshi Hirahara, Toshinori Nakayama
    PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 111(35) 12829-12834 2014年9月  査読有り
    Epigenetic modifications, such as posttranslational modifications of histones, play an important role in gene expression and regulation. These modifications are in part mediated by the Trithorax group (TrxG) complex and the Polycomb group (PcG) complex, which activate and repress transcription, respectively. We herein investigate the role of Menin, a component of the TrxG complex in T helper (Th) cell differentiation and show a critical role for Menin in differentiation and maintenance of Th17 cells. Menin(-/-) T cells do not efficiently differentiate into Th17 cells, leaving Th1 and Th2 cell differentiation intact in in vitro cultures. Menin deficiency resulted in the attenuation of Th17-induced airway inflammation. In differentiating Th17 cells, Menin directly bound to the Il17a gene locus and was required for the deposition of permissive histone modifications and recruitment of the RNA polymerase II transcriptional complex. Interestingly, although Menin bound to the Rorc locus, Menin was dispensable for the induction of Rorc expression and permissive histone modifications in differentiating Th17 cells. In contrast, Menin was required to maintain expression of Rorc in differentiated Th17 cells, indicating that Menin is essential to stabilize expression of the Rorc gene. Thus, Menin orchestrates Th17 cell differentiation and function by regulating both the induction and maintenance of target gene expression.
  • Scott M. Steward-Tharp, Arian Laurence, Yuka Kanno, Alex Kotlyar, Alejandro V. Villarino, Giuseppe Sciume, Stefan Kuchen, Wolfgang Resch, Elizabeth A. Wohlfert, Kan Jiang, Kiyoshi Hirahara, Golnaz Vahedi, Hong-wei Sun, Lionel Feigenbaum, Joshua D. Milner, Steven M. Holland, Rafael Casellas, Fiona Powrie, John J. O'Shea
    BLOOD 123(19) 2978-2987 2014年5月  査読有り
    Mutations of STAT3 underlie the autosomal dominant form of hyperimmunoglobulin E syndrome (HIES). STAT3 has critical roles in immune cells and thus, hematopoietic stem cell transplantation (HSCT), might be a reasonable therapeutic strategy in this disease. However, STAT3 also has critical functions in nonhematopoietic cells and dissecting the protean roles of STAT3 is limited by the lethality associated with germline deletion of Stat3. Thus, predicting the efficacy of HSCT for HIES is difficult. To begin to dissect the importance of STAT3 in hematopoietic and nonhematopoietic cells as it relates to HIES, we generated a mouse model of this disease. We found that these transgenic mice recapitulate multiple aspects of HIES, including elevated serum IgE and failure to generate Th17 cells. We found that these mice were susceptible to bacterial infection that was partially corrected by HSCT using wild-type bone marrow, emphasizing the role played by the epithelium in the pathophysiology of HIES.
  • Shingo Nakayamada, Amanda C. Poholek, Kristina T. Lu, Hayato Takahashi, Masanari Kato, Shigeru Iwata, Kiyoshi Hirahara, Jennifer L. Cannons, Pamela L. Schwartzberg, Golnaz Vahedi, Hong-wei Sun, Yuka Kanno, John J. O'Shea
    JOURNAL OF IMMUNOLOGY 192(5) 2156-2166 2014年3月  査読有り
    CD4(+) T follicular helper cells (TFH) are critical for the formation and function of B cell responses to infection or immunization, but also play an important role in autoimmunity. The factors that contribute to the differentiation of this helper cell subset are incompletely understood, although several cytokines including IL-6, IL-21, and IL-12 can promote TFH cell formation. Yet, none of these factors, nor their downstream cognate STATs, have emerged as nonredundant, essential drivers of TFH cells. This suggests a model in which multiple factors can contribute to the phenotypic characteristics of TFH cells. Because type I IFNs are often generated in immune responses, we set out to investigate whether these factors are relevant to TFH cell differentiation. Type I IFNs promote Th1 responses, thus one possibility was these factors antagonized TFH-expressed genes. However, we show that type I IFNs (IFN-alpha/beta) induced B cell lymphoma 6 (Bcl6) expression, the master regulator transcription factor for TFH cells, and CXCR5 and programmed cell death-1 (encoded by Pdcd1), key surface molecules expressed by TFH cells. In contrast, type I IFNs failed to induce IL-21, the signature cytokine for TFH cells. The induction of Bcl6 was regulated directly by STAT1, which bound to the Bcl6, Cxcr5, and Pdcd1 loci. These data suggest that type I IFNs (IFN-alpha/beta) and STAT1 can contribute to some features of TFH cells but are inadequate in inducing complete programming of this subset.
  • Yusuke Endo, Kiyoshi Hirahara, Ryoji Yagi, Damon J. Tumes, Toshinori Nakayama
    TRENDS IN IMMUNOLOGY 35(2) 69-78 2014年2月  査読有り
    Immunological memory is a hallmark of adaptive immunity. Memory CD4 T helper (Th) cells are central to acquired immunity, and vaccines for infectious diseases are developed based on this concept. However, memory Th cells also play a critical role in the pathogenesis of various chronic inflammatory diseases, including asthma. We refer to these populations as 'pathogenic memory Th cells.' Here, we review recent developments highlighting the functions and characteristics of several pathogenic memory type Th2 cell subsets in allergic inflammation. Also discussed are the similarities and differences between pathogenic memory Th2 cells and recently identified type 2 innate lymphoid cells (ILC2), focusing on cytokine production and phenotypic profiles.
  • Michael Bonelli, Han-Yu Shih, Kiyoshi Hirahara, Kentner Singelton, Arian Laurence, Amanda Poholek, Tim Hand, Yohei Mikami, Golnaz Vahedi, Yuka Kanno, John J. O'Shea
    TRANSCRIPTIONAL CONTROL OF LINEAGE DIFFERENTIATION IN IMMUNE CELLS 381 279-326 2014年  査読有り
    CD4(+) helper T cells are crucial for autoimmune and infectious diseases; however, the recognition of the many, diverse fates available continues unabated. Precisely what controls specification of helper T cells and preserves phenotypic commitment is currently intensively investigated. In this review, we will discuss the major factors that impact helper T cell fate choice, ranging from cytokines and the microbiome to metabolic control and epigenetic regulation. We will also discuss the technological advances along with the attendant challenges presented by "big data," which allow the understanding of these processes on comprehensive scales.
  • Rahul Roychoudhuri, Kiyoshi Hirahara, Kambiz Mousavi, David Clever, Christopher A. Klebanoff, Michael Bonelli, Giuseppe Sciume, Hossein Zare, Golnaz Vahedi, Barbara Dema, Zhiya Yu, Hui Liu, Hayato Takahashi, Mahadev Rao, Pawel Muranski, Joseph G. Crompton, George Punkosdy, Davide Bedognetti, Ena Wang, Victoria Hoffmann, Juan Rivera, Francesco M. Marincola, Atsushi Nakamura, Vittorio Sartorelli, Yuka Kanno, Luca Gattinoni, Akihiko Muto, Kazuhiko Igarashi, John J. O'Shea, Nicholas P. Restifo
    NATURE 498(7455) 506-+ 2013年6月  査読有り
    Through their functional diversification, distinct lineages of CD4(+) T cells can act to either drive or constrain immune-mediated pathology. Transcription factors are critical in the generation of cellular diversity, and negative regulators antagonistic to alternate fates often act in conjunction with positive regulators to stabilize lineage commitment(1). Genetic polymorphisms within a single locus encoding the transcription factor BACH2 are associated with numerous autoimmune and allergic diseases including asthma(2), Crohn's disease(3,4), coeliac disease(5), vitiligo(6), multiple sclerosis(7) and type 1 diabetes(8). Although these associations point to a shared mechanism underlying susceptibility to diverse immune-mediated diseases, a function for BACH2 in the maintenance of immune homeostasis has not been established. Here, by studying mice in which the Bach2 gene is disrupted, we define BACH2 as a broad regulator of immune activation that stabilizes immunoregulatory capacity while repressing the differentiation programs of multiple effector lineages in CD4(+) T cells. BACH2 was required for efficient formation of regulatory (T-reg) cells and consequently for suppression of lethal inflammation in a manner that was T-reg-cell-dependent. Assessment of the genome-wide function of BACH2, however, revealed that it represses genes associated with effector cell differentiation. Consequently, its absence during T-reg polarization resulted in inappropriate diversion to effector lineages. In addition, BACH2 constrained full effector differentiation within T(H)1, T(H)2 and T(H)17 cell lineages. These findings identify BACH2 as a key regulator of CD4(+) T-cell differentiation that prevents inflammatory disease by controlling the balance between tolerance and immunity.
  • Hirahara Kiyoshi, Poholek Amanda, Vahedi Golnaz, Laurence Arian, Kanno Yuka, Milner Joshua D, O'Shea John J
    JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY 131(5) 1276-1287 2013年5月  査読有り
  • Golnaz Vahedi, Amanda C. Poholek, Timothy W. Hand, Arian Laurence, Yuka Kanno, John J. O'Shea, Kiyoshi Hirahara
    IMMUNOLOGICAL REVIEWS 252 24-40 2013年3月  査読有り
    CD4(+) T cells are critical for the elimination of an immense array of microbial pathogens. Among the ways they accomplish this task is to generate progeny with specialized, characteristic patterns of gene expression. From this perspective, helper cells can be viewed as pluripotent precursors that adopt distinct cell fates. Although there are aspects of helper cell differentiation that can be modeled as a classic cell fate commitment, CD4(+) T cells also maintain considerable flexibility in their transcriptional program. This makes sense in terms of host defense, but raises the question of how these remarkable cells balance both these requirements, a high degree of specific gene expression and the capacity for plasticity. In this review, we discuss recent advances in our understanding of CD4(+) T-cell specification, focusing on how genomic perspectives have influenced our views of these processes. The relative contributions of sensors of the cytokine milieu, especially the signal transducer and activator of transcription family transcription factors, 'master regulators', and other transcription factors are considered as they relate to the helper cell transcriptome and epigenome.
  • Hosokawa, H., Tanaka, T., Kato, M., Shinoda, K., Tohyama, H., Hanazawa, A., Tamaki, Y., Hirahara, K., Yagi, R., Sakikawa, I., Morita, A., Nagira, M., Poyurovsky, M.V., Suzuki, Y., Motohashi, S., Nakayama, T.
    Proceedings of the National Academy of Sciences of the United States of America 110(46) 18626-18631 2013年  査読有り
  • Giuseppe Sciume, Kiyoshi Hirahara, Hayato Takahashi, Arian Laurence, Alejandro V. Villarino, Kentner L. Singleton, Sean P. Spencer, Christoph Wilhelm, Amanda C. Poholek, Golnaz Vahedi, Yuka Kanno, Yasmine Belkaid, John J. O'Shea
    JOURNAL OF EXPERIMENTAL MEDICINE 209(13) 2331-2338 2012年12月  査読有り
    Interleukin (IL)-22-producing innate lymphoid cells (ILCs; ILC22) comprise a heterogeneous population of cells that are dependent on the transcription factor retinoid-related orphan gamma t (ROR gamma t) and are critical for barrier function of the intestinal mucosa. A distinct ILC22 subset expresses the natural cytotoxicity receptor NKp46 (NKp46(+) ILC22); however, the factors that contribute to the generation of this population versus other subsets are largely unknown. Herein, we show that T-bet (encoded by Tbx21) was highly expressed in NKp46(+) ILC22, a feature shared by all NKp46(+) cells present in the intestine but not by other IL-22-producing populations. Accordingly, the absence of T-bet resulted in loss of NKp46+ ILC22 in the intestinal lamina propria. The residual NKp46(+) ILC22 present in Tbx21(-/-) mice showed a marked reduction of Ror gamma t expression and impairment in IL-22 production. Generation and functions of gut NK1.1(+) cells were also altered. Bone marrow chimera experiments revealed a cell-intrinsic requirement for T-bet in these subsets and competitive reconstitution experiments revealed roles for T-bet in multiple ILC subsets. Thus, T-bet has a general importance for ILC in the gut and plays a selective and critical role in the generation of NKp46(+) ILC22.
  • Hayato Takahashi, Tomohiko Kanno, Shingo Nakayamada, Kiyoshi Hirahara, Giuseppe Sciume, Stefan A. Muljo, Stefan Kuchen, Rafael Casellas, Lai Wei, Yuka Kanno, John J. O'Shea
    NATURE IMMUNOLOGY 13(6) 587-+ 2012年6月  査読有り
    Distinct CD4(+) T cell subsets are critical for host defense and immunoregulation. Although these subsets can act as terminally differentiated lineages, they have been increasingly noted to demonstrated plasticity. MicroRNAs are factors that control T cell stability and plasticity. Here we report that naturally occurring regulatory T cells (T-reg cells) had high expression of the microRNA miR-10a and that miR-10a was induced by retinoic acid and transforming growth factor-beta (TGF-beta) in inducible T-reg cells. By simultaneously targeting the transcriptional repressor Bcl-6 and the corepressor Ncor2, miR-10a attenuated the phenotypic conversion of inducible T-reg cells into follicular helper T cells. We also found that miR-10a limited differentiation into the T(H)17 subset of helper T cells and therefore represents a factor that can fine-tune the plasticity and fate of helper T cells.
  • Kiyoshi Hirahara, Kamran Ghoreschi, Xiang-Ping Yang, Hayato Takahashi, Arian Laurence, Golnaz Vahedi, Giuseppe Sciume, Aisling O'Hara Hall, Christopher D. Dupont, Loise M. Francisco, Qian Chen, Masao Tanaka, Yuka Kanno, Hong-Wei Sun, Arlene H. Sharpe, Christopher A. Hunter, John J. O'Shea
    IMMUNITY 36(6) 1017-1030 2012年6月  査読有り
    Interleukin-27 (IL-27) is a key immunosuppressive cytokine that counters T helper 17 (Th17) cell-mediated pathology. To identify mechanisms by which IL-27 might exert its immunosuppressive effect, we analyzed genes in T cells rapidly induced by IL-27. We found that IL-27 priming of naive T cells upregulated expression of programmed death ligand 1 (PD-L1) in a signal transducer and activator of transcription 1 (STAT1)-dependent manner. When cocultured with naive CD4(+) T cells, IL-27-primed T cells inhibited the differentiation of Th17 cells in trans through a PD-1-PD-L1 interaction. In vivo, coadministration of naive TCR transgenic T cells (2D2 T cells) with IL-27-primed T cells expressing PD-L1 inhibited the development of Th17 cells and protected from severe autoimmune encephalomyelitis. Thus, these data identify a suppressive activity of IL-27, by which CD4(+) T cells can restrict differentiation of Th17 cells in trans.
  • Kiyoshi Hirahara, Kamran Ghoreschi, Xiang-Ping Yang, Hayato Takahashi, Arian Laurence, Golnaz Vahedi, Giuseppe Sciume, Aisling Hall, Christopher Dupont, Loise Francisco, Qian Chen, Masao Tanaka, Yuka Kanno, Hong-Wei Sun, Arlene Sharpe, Christopher Hunter, John O'Shea
    JOURNAL OF IMMUNOLOGY 188 2012年5月  査読有り
  • Yuka Kanno, Golnaz Vahedi, Kiyoshi Hirahara, Kentner Singleton, John J. O'Shea
    ANNUAL REVIEW OF IMMUNOLOGY, VOL 30 30 707-731 2012年  査読有り
    T helper cell differentiation occurs in the context of the extracellular cytokine milieu evoked by diverse microbes and other pathogenic stimuli along with T cell receptor stimulation. The culmination of these signals results in specification of T helper lineages, which occurs through the combinatorial action of multiple transcription factors that establish distinctive transcriptomes. In this manner, inducible, but constitutively active, master regulators work in conjunction with factors such as the signal transducer and activator of transcriptions (STATs) that sense the extracellular environment. The acquisition of a distinctive transcriptome also depends on chromatin modifications that impact key cis elements as well as the changes in global genomic organization. Thus, signal transduction and epigenetics are linked in these processes of differentiation. In this review, recent advances in understanding T helper lineage specification and deciphering the action of transcription factors are summarized with emphasis on comprehensive views of the dynamic T cell epigenome.
  • Laurence, A, Ghoreschi, K, Hirahara, K, Yang, X. P, O’Shea, J. J
    Inflammation and Regeneration 32(1) 16-22 2012年  査読有り
  • Kiyoshi Hirahara, Golnaz Vahedi, Kamran Ghoreschi, Xiang-Ping Yang, Shingo Nakayamada, Yuka Kanno, John J. O&apos, Shea, Arian Laurence
    IMMUNOLOGY 134(3) 235-245 2011年11月  査読有り
    CD4(+) T cells have critical roles in orchestrating immune responses to diverse microbial pathogens. This is accomplished through the differentiation of CD4(+) T helper cells to specialized subsets in response to microbial pathogens, which evoke a distinct cytokine milieu. Signal transducer and activator of transcription family transcription factors sense these cytokines and they in turn regulate expression of lineage-defining master regulators that programme selective gene expression, resulting in distinctive phenotypes. However, phenotype and restricted gene expression are determined not only by the action of transcription factors; chromatin accessibility is required for these factors to exert their effect. Technical advances have greatly expanded our understanding of transcription factor action and dynamic changes in the epigenome that accompany cellular differentiation. In this review, we will discuss recent progress in the understanding of how cytokines influence gene expression and epigenetic modifications, and the impact of these findings on our views of helper cell lineage commitment and plasticity.
  • Kamran Ghoreschi, Arian Laurence, Xiang-Ping Yang, Kiyoshi Hirahara, John J. O&apos, Shea
    TRENDS IN IMMUNOLOGY 32(9) 395-401 2011年9月  査読有り
    T helper (Th)17 cells have been proposed to represent a new CD4(+) T cell lineage that is important for host defense against fungi and extracellular bacteria, and the development of autoimmune diseases. Precisely how these cells arise has been the subject of some debate, with apparent species-specific differences in mice and humans. Here, we describe evolving views of Th17 specification, highlighting the contribution of transforming growth factor-beta and the opposing roles of signal transducer and activator of transcription (STAT)3 and STAT5. Increasing evidence points to heterogeneity and inherent phenotypic instability in this subset. Ideally, better understanding of expression and action of key transcription factors and the epigenetic landscape of Th17 can help explain the flexibility and diversity of interleukin-17-producing cells.
  • Xiang-Ping Yang, Kamran Ghoreschi, Scott M. Steward-Tharp, Jaime Rodriguez-Canales, Jinfang Zhu, John R. Grainger, Kiyoshi Hirahara, Hong-Wei Sun, Lai Wei, Golnaz Vahedi, Yuka Kanno, John J. O&apos, Shea, Arian Laurence
    NATURE IMMUNOLOGY 12(3) 247-U84 2011年3月  査読有り
    Interleukin 2 (IL-2), a cytokine linked to human autoimmune disease, limits IL-17 production. Here we found that deletion of the gene encoding the transcription factor STAT3 in T cells abrogated IL-17 production and attenuated autoimmunity associated with IL-2 deficiency. Whereas STAT3 induced IL-17 and the transcription factor ROR gamma t and inhibited the transcription factor Foxp3, IL-2 inhibited IL-17 independently of Foxp3 and ROR gamma t. STAT3 and STAT5 bound to multiple common sites across the locus encoding IL-17. The induction of STAT5 binding by IL-2 was associated with less binding of STAT3 at these sites and the inhibition of associated active epigenetic marks. &apos;Titration&apos; of the relative activation of STAT3 and STAT5 modulated the specification of cells to the IL-17-producing helper T cell (T(H)17 cell) subset. Thus, the balance rather than the absolute magnitude of these signals determined the propensity of cells to make a key inflammatory cytokine.
  • Kiyoshi Hirahara, Kamran Ghoreschi, Arian Laurence, Xiang-Ping Yang, Yuka Kanno, John J. O&apos, Shea
    CYTOKINE & GROWTH FACTOR REVIEWS 21(6) 425-434 2010年12月  査読有り
    Over the last decade, our understanding of helper/effector T cell differentiation has changed dramatically. The discovery of interleukin (IL-)17-producing T cells (Th17) and other subsets has changed our view of T cell-mediated immunity. Characterization of the signaling pathways involved in the Th17 commitment has provided exciting new insights into the differentiation of CD4(+) T cells. Importantly, the emerging data on conversion among polarized T helper cells have raised the question how we should view such concepts as T cell lineage commitment, terminal differentiation and plasticity. In this review, we will discuss the current understanding of the signaling pathways, molecular interactions, and transcriptional and epigenetic events that contribute to Th17 differentiation and acquisition of effector functions. Published by Elsevier Ltd.
  • Toshinori Nakayama, Kiyoshi Hirahara, Masakatsu Yamasahita
    JOURNAL OF IMMUNOLOGY 182 2009年4月  査読有り
  • Kiyoshi Hirahara, Masakatsu Yamashita, Chiaki Iwamura, Kenta Shinoda, Akihiro Hasegawa, Hirohisa Yoshizawa, Haruhiko Koseki, Fumitake Gejyo, Toshinori Nakayama
    JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY 122(3) 512-520 2008年9月  査読有り
    Background: Studies of human asthma and of animal models of allergic inflammation/asthma highlight a crucial role for T(H)2 cells in the pathogenesis of allergic asthma. Repressor of GATA (ROG) is a POZ (BTB) domain-containing Kruppel-type zinc finger family (or POK family) repressor. A repressive function to GATA3, a master transcription factor for T(H)2 cell differentiation, is indicated. Objective: The aim of this study was to clarify the regulatory roles of ROG in the pathogenesis of T(H)2-driven allergic diseases, such as allergic asthma. Methods: We examined allergic airway inflammation and airway hyperresponsiveness (AHR) in 3 different mouse models, which use either ROG-deficient (ROG(-/-)) mice, ROG transgenic mice, or adoptive transfer of cells. Results: In ROG(-/-) mice T(H)2 cell differentiation, T(H)2 responses, eosinophilic airway inflammation, and AHR were enhanced. In ROG transgenic mice the levels of eosinophilic airway inflammation and AHR were dramatically reduced. Furthermore, adoptive transfer of T(H)2 cells with increased or decreased levels of ROG expression into the asthmatic mice resulted in reduced or enhanced airway inflammation, respectively. Conclusion: These results indicate that ROG regulates allergic airway inflammation and AHR in a negative manner, and thus ROG might represent another potential therapeutic target for the treatment of asthmatic patients.
  • Yamashita, M., Kuwahara, M., Suzuki, A., Hirahara, K., Shinnaksu, R., Hosokawa, H., Hasegawa, A., Motohashi, S., Iwama, A., Nakayama, T.
    Journal of Experimental Medicine 205(5) 1109-1120 2008年  査読有り
  • YI Nigo, M Yamashita, K Hirahara, R Shinnakasu, M Inami, M Kimura, A Hasegawa, Y Kohno, T Nakayama
    PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 103(7) 2286-2291 2006年2月  査読有り
    In a mouse experimental asthma model, the administration of bacterial lipopolysaccharide (LIPS), particularly at low doses, enhances the levels of ovalbumin (OVA)-induced eosinophilic airway inflammation. In an effort to clarify the cellular and molecular basis for the LPS effect, we demonstrate that the OVA-induced eosinophilic inflammation in the lung is dramatically increased by the administration of LPS in wild-type mice, whereas such increase was not observed in mast-cell-deficient mice or Toll-like receptor (TLR)4-deficient mice. Adoptive transfer of bone-marrow-derived mast cells (BMMCs) from wild-type, but not from TLR4-deficient, mice restored the increased eosinophilic inflammation in mast-cell-deficient mice. Wild-type BMMCs pretreated with LPS in vitro also reconstituted the eosinophilic inflammation. Moreover, in vitro analysis revealed that the treatment of BMMCs with LPS resulted in NF-kappa B activation, sustained up-regulation of GATA1 and -2 expression, and increased the capability to produce IL-5 and -13. Dramatic increases in the expression of IL-5 and -13 and Eotaxin 2 were detected in LIPS-treated BMMCs after costimulation with LIPS and IgE/Ag. Overexpression of GATA1, but not GATA2, in MC9 mast cells resulted in increased transcriptional activity of IL-4, -5, and -13. Furthermore, the levels of transcription of Th2 cytokines in BMMCs were decreased by the introduction of small interfering RNA for GATA1. Thus, mast cells appear to control allergic airway inflammation after their activation and modulation through TLR4-mediated induction of GATA1 and subsequent increase in Th2 cytokine production.
  • Yamashita, M., Hirahara, K., Shinnakasu, R., Hosokawa, H., Norikane, S., Kimura, M.Y., Hasegawa, A., Nakayama, T.
    Immunity 24(5) 611-622 2006年  査読有り

MISC

 59

書籍等出版物

 5

講演・口頭発表等

 152

共同研究・競争的資金等の研究課題

 15

産業財産権

 1