Network


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

Hotspot


Dive into the research topics where Yuka Kanno is active.

Publication


Featured researches published by Yuka Kanno.


Nature | 2008

Impaired T H 17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome

Joshua D. Milner; Jason M. Brenchley; Arian Laurence; Alexandra F. Freeman; Brenna J. Hill; Kevin M. Elias; Yuka Kanno; Christine Spalding; Houda Elloumi; Michelle L. Paulson; Joie Davis; Amy P. Hsu; Ava I. Asher; John J. O'Shea; Steven M. Holland; William E. Paul

The autosomal dominant hyper-IgE syndrome (HIES, ‘Job’s syndrome’) is characterized by recurrent and often severe pulmonary infections, pneumatoceles, eczema, staphylococcal abscesses, mucocutaneous candidiasis, and abnormalities of bone and connective tissue. Mutations presumed to underlie HIES have recently been identified in stat3, the gene encoding STAT3 (signal transducer and activator of transcription 3) (refs 3, 4). Although impaired production of interferon-γ and tumour-necrosis factor by T cells, diminished memory T-cell populations, decreased delayed-type-hypersensitivity responses and decreased in vitro lymphoproliferation in response to specific antigens have variably been described, specific immunological abnormalities that can explain the unique susceptibility to particular infections seen in HIES have not yet been defined. Here we show that interleukin (IL)-17 production by T cells is absent in HIES individuals. We observed that ex vivo T cells from subjects with HIES failed to produce IL-17, but not IL-2, tumour-necrosis factor or interferon-γ, on mitogenic stimulation with staphylococcal enterotoxin B or on antigenic stimulation with Candida albicans or streptokinase. Purified naive T cells were unable to differentiate into IL-17-producing (TH17) T helper cells in vitro and had lower expression of retinoid-related orphan receptor (ROR)-γt, which is consistent with a crucial role for STAT3 signalling in the generation of TH17 cells. TH17 cells have emerged as an important subset of helper T cells that are believed to be critical in the clearance of fungal and extracellular bacterial infections. Thus, our data suggest that the inability to produce TH17 cells is a mechanism underlying the susceptibility to the recurrent infections commonly seen in HIES.


Immunity | 2009

Global Mapping of H3K4me3 and H3K27me3 Reveals Specificity and Plasticity in Lineage Fate Determination of Differentiating CD4+ T Cells

Gang Wei; Lai Wei; Chongzhi Zang; Jane Hu-Li; Zhengju Yao; Kairong Cui; Yuka Kanno; Tae-Young Roh; Wendy T. Watford; Dustin E. Schones; Weiqun Peng; Hong-Wei Sun; William E. Paul; John J. O'Shea; Keji Zhao

Multipotential naive CD4(+) T cells differentiate into distinct lineages including T helper 1 (Th1), Th2, Th17, and inducible T regulatory (iTreg) cells. The remarkable diversity of CD4(+) T cells begs the question whether the observed changes reflect terminal differentiation with heritable epigenetic modifications or plasticity in T cell responses. We generated genome-wide histone H3 lysine 4 (H3K4) and lysine 27 (H3K27) trimethylation maps in naive, Th1, Th2, Th17, iTreg, and natural Treg (nTreg) cells. We found that although modifications of signature-cytokine genes (Ifng, Il4, and Il17) partially conform to the expectation of lineage commitment, genes encoding transcription factors like Tbx21 exhibit a broad spectrum of epigenetic states, consistent with our demonstration of T-bet and interferon-gamma induction in nTreg cells. Our data suggest an epigenetic mechanism underlying the specificity and plasticity of effector and regulatory T cells and also provide a framework for understanding complexity of CD4(+) T helper cell differentiation.


Nature | 2010

Generation of pathogenic T(H)17 cells in the absence of TGF-β signalling.

Kamran Ghoreschi; Arian Laurence; Xiang-Ping Yang; Cristina M. Tato; Mandy J. McGeachy; Joanne E. Konkel; Haydeé L. Ramos; Lai Wei; Todd S. Davidson; Nicolas Bouladoux; John Grainger; Qian Chen; Yuka Kanno; Wendy T. Watford; Hong-Wei Sun; Gérard Eberl; Ethan M. Shevach; Yasmine Belkaid; Daniel J. Cua; WanJun Chen; John J. O’Shea

CD4+ T-helper cells that selectively produce interleukin (IL)-17 (TH17), are critical for host defence and autoimmunity. Although crucial for TH17 cells in vivo, IL-23 has been thought to be incapable of driving initial differentiation. Rather, IL-6 and transforming growth factor (TGF)-β1 have been proposed to be the factors responsible for initiating specification. Here we show that TH17 differentiation can occur in the absence of TGF-β signalling. Neither IL-6 nor IL-23 alone efficiently generated TH17 cells; however, these cytokines in combination with IL-1β effectively induced IL-17 production in naive precursors, independently of TGF-β. Epigenetic modification of the Il17a, Il17f and Rorc promoters proceeded without TGF-β1, allowing the generation of cells that co-expressed RORγt (encoded by Rorc) and T-bet. T-bet+RORγt+ TH17 cells are generated in vivo during experimental allergic encephalomyelitis, and adoptively transferred TH17 cells generated with IL-23 without TGF-β1 were pathogenic in this disease model. These data indicate an alternative mode for TH17 differentiation. Consistent with genetic data linking IL23R with autoimmunity, our findings re-emphasize the importance of IL-23 and therefore may have therapeutic implications.


Cell | 1996

Immunodeficiency and Chronic Myelogenous Leukemia-like Syndrome in Mice with a Targeted Mutation of the ICSBP Gene

Thomas Holtschke; Jürgen Löhler; Yuka Kanno; Thomas Fehr; Nathalia Giese; Frank Rosenbauer; Jing Lou; Klaus-Peter Knobeloch; Lucia Gabriele; Jeffrey F. Waring; Martin F. Bachmann; Rolf M. Zinkernagel; Herbert C. Morse; Keiko Ozato; Ivan Horak

Interferon consensus sequence binding protein (ICSBP) is a transcription factor of the interferon (IFN) regulatory factor (IRF) family. Mice with a null mutation of ICSBP exhibit two prominent phenotypes related to previously described activities of the IRF family. The first is enhanced susceptibility to virus infections associated with impaired production of IFN(gamma). The second is deregulated hematopoiesis in both ICSBP-/- and ICSBP+/- mice that manifests as a syndrome similar to human chronic myelogenous leukemia. The chronic period of the disease progresses to a fatal blast crisis characterized by a clonal expansion of undifferentiated cells. Normal mice injected with cells from mice in blast crisis developed acute leukemia within 6 weeks of transfer. These results suggest a novel role for ICSBP in regulating the proliferation and differentiation of hematopoietic progenitor cells.


Journal of Experimental Medicine | 2009

Lymphoid tissue inducer–like cells are an innate source of IL-17 and IL-22

Hiroaki Takatori; Yuka Kanno; Wendy T. Watford; Cristina M. Tato; Greta Weiss; Ivaylo I. Ivanov; Dan R. Littman; John J. O'Shea

The interleukin (IL) 17 family of cytokines has emerged to be critical for host defense as well as the pathogenesis of autoimmune and autoinflammatory disorders, and serves to link adaptive and innate responses. Recent studies have identified a new subset of T cells that selectively produce IL-17 (Th17 cells; Bettelli, E., T. Korn, and V.K. Kuchroo. 2007. Curr. Opin. Immunol. 19:652–657; Kolls, J.K., and A. Linden. 2004. Immunity. 21:467–476), but the regulation of IL-17 production by innate immune cells is less well understood. We report that in vitro stimulation with IL-23 induced IL-17 production by recombination activating gene (Rag) 2−/− splenocytes but not Rag2−/− common γ chain−/− splenocytes. We found that a major source of IL-17 was CD4+CD3−NK1.1−CD11b−Gr1−CD11c−B220− cells, a phenotype that corresponds to lymphoid tissue inducer–like cells (LTi-like cells), which constitutively expressed the IL-23 receptor, aryl hydrocarbon receptor, and CCR6. In vivo challenge with the yeast cell wall product zymosan rapidly induced IL-17 production in these cells. Genetic deletion of signal transducer and activator of transcription 3 reduced but did not abrogate IL-17 production in LTi-like cells. Thus, it appears that splenic LTi-like cells are a rapid source of IL-17 and IL-22, which might contribute to dynamic organization of secondary lymphoid organ structure or host defense.


Immunological Reviews | 2004

Signaling by IL-12 and IL-23 and the immunoregulatory roles of STAT4

Wendy T. Watford; Bruce D. Hissong; Jay H. Bream; Yuka Kanno; Linda M. Muul; John J. O'Shea

Summary:  Produced in response to a variety of pathogenic organisms, interleukin (IL)‐12 and IL‐23 are key immunoregulatory cytokines that coordinate innate and adaptive immune responses. These dimeric cytokines share a subunit, designated p40, and bind to a common receptor chain, IL‐12Rβ1. The receptor for IL‐12 is composed of IL‐12Rβ1 and IL‐12Rβ2, whereas IL‐23 binds to a receptor composed of IL‐12Rβ1 and IL‐23R. Both cytokines activate the Janus kinases Tyk2 and Jak2, the transcription factor signal transducer and activator of transcription 4 (STAT4), as well as other STATs. A major action of IL‐12 is to promote the differentiation of naïve CD4+ T cells into T‐helper (Th) 1 cells, which produce interferon (IFN)‐γ, and deficiency of IL‐12, IL‐12R subunits or STAT4 is similar in many respects. In contrast, IL‐23 promotes end‐stage inflammation. Targeting IL‐12, IL‐23, and their downstream signaling elements would therefore be logical strategies for the treatment of immune‐mediated diseases.


Immunity | 2010

Diverse targets of the transcription factor STAT3 contribute to T cell pathogenicity and homeostasis.

Lydia Durant; Wendy T. Watford; Haydeé L. Ramos; Arian Laurence; Golnaz Vahedi; Lai Wei; Hayato Takahashi; Hong-Wei Sun; Yuka Kanno; Fiona Powrie; John J. O'Shea

STAT3, an essential transcription factor with pleiotropic functions, plays critical roles in the pathogenesis of autoimmunity. Despite recent data linking STAT3 with inflammatory bowel disease, exactly how it contributes to chronic intestinal inflammation is not known. Using a T cell transfer model of colitis, we found that STAT3 expression in T cells was essential for the induction of both colitis and systemic inflammation. STAT3 was critical in modulating the balance of T helper 17 (Th17) and regulatory T (Treg) cells, as well as in promoting CD4(+) T cell proliferation. We used chromatin immunoprecipitation and massive parallel sequencing (ChIP-Seq) to define the genome-wide targets of STAT3 in CD4(+) T cells. We found that STAT3 bound to multiple genes involved in Th17 cell differentiation, cell activation, proliferation, and survival, regulating both expression and epigenetic modifications. Thus, STAT3 orchestrates multiple critical aspects of T cell function in inflammation and homeostasis.


Journal of Experimental Medicine | 2006

T-bet regulates Th1 responses through essential effects on GATA-3 function rather than on IFNG gene acetylation and transcription

Takashi Usui; Jan Preiss; Yuka Kanno; Zheng Ju Yao; Jay H. Bream; John J. O'Shea; Warren Strober

T helper type 1 (Th1) development is facilitated by interrelated changes in key intracellular factors, particularly signal transducer and activator of transcription (STAT)4, T-bet, and GATA-3. Here we show that CD4+ cells from T-bet−/− mice are skewed toward Th2 differentiation by high endogenous GATA-3 levels but exhibit virtually normal Th1 differentiation provided that GATA-3 levels are regulated at an early stage by anti–interleukin (IL)-4 blockade of IL-4 receptor (R) signaling. In addition, under these conditions, Th1 cells from T-bet−/− mice manifest IFNG promotor accessibility as detected by histone acetylation and deoxyribonuclease I hypersensitivity. In related studies, we show that the negative effect of GATA-3 on Th1 differentiation in T-bet−/− cells arises from its ability to suppress STAT4 levels, because if this is prevented by a STAT4-expressing retrovirus, normal Th1 differentiation is observed. Finally, we show that retroviral T-bet expression in developing and established Th2 cells leads to down-regulation of GATA-3 levels. These findings lead to a model of T cell differentiation that holds that naive T cells tend toward Th2 differentiation through induction of GATA-3 and subsequent down-regulation of STAT4/IL-12Rβ2 chain unless GATA-3 levels or function is regulated by T-bet. Thus, the principal function of T-bet in developing Th1 cells is to negatively regulate GATA-3 rather than to positively regulate the IFNG gene.


Molecular Cell | 2004

Selective Recognition of Acetylated Histones by Bromodomain Proteins Visualized in Living Cells

Tomohiko Kanno; Yuka Kanno; Richard M. Siegel; Moon Kyoo Jang; Michael J. Lenardo; Keiko Ozato

Acetylation and other modifications on histones comprise histone codes that govern transcriptional regulatory processes in chromatin. Yet little is known how different histone codes are translated and put into action. Using fluorescence resonance energy transfer, we show that bromodomain-containing proteins recognize different patterns of acetylated histones in intact nuclei of living cells. The bromodomain protein Brd2 selectively interacted with acetylated lysine 12 on histone H4, whereas TAF(II)250 and PCAF recognized H3 and other acetylated histones, indicating fine specificity of histone recognition by different bromodomains. This hierarchy of interactions was also seen in direct peptide binding assays. Interaction with acetylated histone was essential for Brd2 to amplify transcription. Moreover association of Brd2, but not other bromodomain proteins, with acetylated chromatin persisted on chromosomes during mitosis. Thus the recognition of histone acetylation code by bromodomains is selective, is involved in transcription, and potentially conveys transcriptional memory across cell divisions.


Immunity | 2011

Early Th1 Cell Differentiation Is Marked by a Tfh Cell-like Transition

Shingo Nakayamada; Yuka Kanno; Hayato Takahashi; Dragana Jankovic; Kristina T. Lu; Thomas A. Johnson; Hong-Wei Sun; Golnaz Vahedi; Ofir Hakim; Robin Handon; Pamela L. Schwartzberg; Gordon L. Hager; John J. O'Shea

Follicular helper T (Tfh) cells comprise an important subset of helper T cells; however, their relationship with other helper lineages is incompletely understood. Herein, we showed interleukin-12 acting via the transcription factor STAT4 induced both Il21 and Bcl6 genes, generating cells with features of both Tfh and Th1 cells. However, STAT4 also induced the transcription factor T-bet. With ChIP-seq, we defined the genome-wide targets of T-bet and found that it repressed Bcl6 and other markers of Tfh cells, thereby attenuating the nascent Tfh cell-like phenotype in the late phase of Th1 cell specification. Tfh-like cells were rapidly generated after Toxoplasma gondii infection in mice, but T-bet constrained Tfh cell expansion and consequent germinal center formation and antibody production. Our data argue that Tfh and Th1 cells share a transitional stage through the signal mediated by STAT4, which promotes both phenotypes. However, T-bet represses Tfh cell functionalities, promoting full Th1 cell differentiation.

Collaboration


Dive into the Yuka Kanno's collaboration.

Top Co-Authors

Avatar

John J. O'Shea

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Golnaz Vahedi

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

John J. O’Shea

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Arian Laurence

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hong-Wei Sun

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Giuseppe Sciumè

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Keiko Ozato

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge