Network


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

Hotspot


Dive into the research topics where Yukino Chiba is active.

Publication


Featured researches published by Yukino Chiba.


Clinical & Developmental Immunology | 2010

Regulation of Antitumor Immune Responses by the IL-12 Family Cytokines, IL-12, IL-23, and IL-27

Mingli Xu; Izuru Mizoguchi; Noriko Morishima; Yukino Chiba; Junichiro Mizuguchi; Takayuki Yoshimoto

The interleukin (IL)-12 family, which is composed of heterodimeric cytokines including IL-12, IL-23, and IL-27, is produced by antigen-presenting cells such as macrophages and dendritic cells and plays critical roles in the regulation of helper T (Th) cell differentiation. IL-12 induces IFN-γ production by NK and T cells and differentiation to Th1 cells. IL-23 induces IL-17 production by memory T cells and expands and maintains inflammatory Th17 cells. IL-27 induces the early Th1 differentiation and generation of IL-10-producing regulatory T cells. In addition, these cytokines induce distinct immune responses to tumors. IL-12 activates signal transducers and activator of transcription (STAT)4 and enhances antitumor cellular immunity through interferon (IFN)-γ production. IL-27 activates STAT1, as does IFN-γ and STAT3 as well, and enhances antitumor immunity by augmenting cellular and humoral immunities. In contrast, although exogenously overexpressed IL-23 enhances antitumor immunity via memory T cells, endogenous IL-23 promotes protumor immunity through STAT3 activation by inducing inflammatory responses including IL-17 production.


BioMed Research International | 2010

A pivotal role for interleukin-27 in CD8+ T cell functions and generation of cytotoxic T lymphocytes.

Noriko Morishima; Izuru Mizoguchi; Masae Okumura; Yukino Chiba; Mingli Xu; Motomu Shimizu; Masanori Matsui; Junichiro Mizuguchi; Takayuki Yoshimoto

Cytotoxic T lymphocytes (CTLs) play a critical role in the control of various cancers and infections, and therefore the molecular mechanisms of CTL generation are a critical issue in designing antitumor immunotherapy and vaccines which augment the development of functional and long-lasting memory CTLs. Interleukin (IL)-27, a member of the IL-6/IL-12 heterodimeric cytokine family, acts on naive CD4+ T cells and plays pivotal roles as a proinflammatory cytokine to promote the early initiation of type-1 helper differentiation and also as an antiinflammatory cytokine to limit the T cell hyperactivity and production of pro-inflammatory cytokines. Recent studies revealed that IL-27 plays an important role in CD8+ T cells as well. Therefore, this article reviews current understanding of the role of IL-27 in CD8+ T cell functions and generation of CTLs.


European Journal of Immunology | 2011

Regulation of the development of acute hepatitis by IL-23 through IL-22 and IL-17 production.

Mingli Xu; Noriko Morishima; Izuru Mizoguchi; Yukino Chiba; Koji Fujita; Masahiko Kuroda; Yoichiro Iwakura; Daniel J. Cua; Koji Yasutomo; Junichiro Mizuguchi; Takayuki Yoshimoto

IL‐23 plays a critical role in the expansion of highly proinflammatory Th17 cells secreting IL‐17 and IL‐22. Recently, we demonstrated that Notch signaling drives IL‐22 secretion through the aryl hydrocarbon receptor (AHR) and plays a protective role in Con A‐induced hepatitis. In this study, we investigated the role of IL‐23 in hepatitis using IL‐23p19‐ and IL‐17‐deficient mice. In WT mice, the injection of Con A induced the upregulation of various cytokines, which included IL‐23, IL‐22, IL‐17, IFN‐γ and TNF‐α. In IL‐23p19‐deficient mice, exacerbated hepatitis was observed and serum IL‐22 and IL‐17 levels were greatly reduced, whereas in IL‐17‐deficient mice, ameliorated hepatitis was observed. The injection of exogenous IL‐22 protected p19‐deficient mice from hepatitis, whereas the injection of exogenous IL‐23 significantly increased the serum levels of not only IL‐22 but also IL‐17, and less effectively protected against hepatitis in IL‐17‐dependent and ‐independent manners. Finally, it was revealed that STAT3, STAT4 and Notch contributed to the production of both the cytokines, and that the AHR was important only for IL‐22 production in response to Con A and IL‐23 in liver mononuclear cells. These results suggest that IL‐23 plays a protective role in hepatitis through IL‐22 production and also a pathological role via IL‐17‐dependent and ‐independent mechanisms.


Immunology Letters | 2011

IL-27 suppresses RANKL expression in CD4+ T cells in part through STAT3.

Sadahiro Kamiya; Masae Okumura; Yukino Chiba; Takeshi Fukawa; Chika Nakamura; Noriyuki Nimura; Junichiro Mizuguchi; Seiki Wada; Takayuki Yoshimoto

The receptor activator of NF-κB ligand (RANKL), which is expressed by not only osteoblasts but also activated T cells, plays an important role in bone-destructive diseases such as rheumatoid arthritis. IL-27, a member of the IL-6/IL-12 family cytokines, activates STAT1 and STAT3, promotes early helper T (Th)1 differentiation and generation of IL-10-producing type 1 regulatory T (Tr1) cells, and suppresses the production of inflammatory cytokines and inhibits Th2 differentiation. In addition, IL-27 was recently demonstrated to not only inhibit Th17 differentiation but also directly act on osteoclast precursor cells and suppress RANKL-mediated osteoclastogenesis through STAT1-dependent inhibition of c-Fos, leading to amelioration of the inflammatory bone destruction. In the present study, we investigated the effect of IL-27 on the expression of RANKL in CD4(+) T cells. We found that IL-27 greatly inhibits cell surface expression of RANKL on naive CD4(+) T cells activated by T cell receptor ligation and secretion of its soluble RANKL as well. The inhibitory effect was mediated in part by STAT3 but not by STAT1 or IL-10. In contrast, in differentiated Th17 cells, IL-27 much less efficiently inhibited the RANKL expression after restimulation. Taken together, these results indicate that IL-27 greatly inhibits primary RANKL expression in CD4(+) T cells, which could contribute to the suppressive effects of IL-27 on the inflammatory bone destruction.


Frontiers in Immunology | 2016

Expanding Diversity in Molecular Structures and Functions of the IL-6/IL-12 Heterodimeric Cytokine Family

Hideaki Hasegawa; Izuru Mizoguchi; Yukino Chiba; Mio Ohashi; Mingli Xu; Takayuki Yoshimoto

The interleukin (IL)-6/IL-12 family cytokines have pleiotropic functions and play critical roles in multiple immune responses. This cytokine family has very unique characteristics in that they comprise two distinct subunits forming a heterodimer and each cytokine and receptor subunit shares with each other. The members of this cytokine family are increasing; currently, there are more than six cytokines, including the tentatively named cytokines IL-Y (p28/p40), IL-12 (p35/p40), IL-23 (p19/p40), IL-27 [p28/Epstein–Barr virus-induced protein 3 (EBI3)], IL-35 (p35/EBI3), and IL-39 (p19/EBI3). This family of cytokines covers a very broad range of immune responses, including pro-inflammatory responses, such as helper T (Th)1, Th2, and Th17, to anti-inflammatory responses, such as regulatory T (Treg) cells and IL-10-producing Treg cells. IL-12 is the first member of this family, and IL-12, IL-23, and IL-27 are mainly produced by activated antigen-presenting cells, such as dendritic cells and macrophages. IL-12 plays a critical role in the promotion of Th1 immune responses by inducing interferon-γ production to combat pathogens and malignant tumors. IL-23 induces IL-17 production and is necessary to maintain pathogenic Th17 cells that cause inflammatory and autoimmune diseases. IL-27 was initially reported to play a critical role in promotion of Th1 differentiation; however, subsequent studies revealed that IL-27 has broader stimulatory and inhibitory roles by inducing IL-10-producing Treg cells. IL-35 is produced by forkhead box P3+ Treg cells and activated B cells and has immunosuppressive functions to maintain immune tolerance. The most recently identified cytokine, IL-39, is produced by activated B cells and has pro-inflammatory functions. The cytokine tentatively named IL-Y seems to have anti-inflammatory functions by inhibiting Th1 and Th17 differentiation. In addition, individual cytokine subunits were also shown to have self-standing activities. Thus, promiscuity within the IL-6/IL-12 family cytokines complicates structural and functional clarification and assignment of individual cytokines. A better understanding of the recent advances and expanding diversity in molecular structures and functions of the IL-6/IL-12 family cytokines could allow the creation of novel therapeutic strategies by using them as tools and targeted molecules.


PLOS Pathogens | 2016

Promotion of Expansion and Differentiation of Hematopoietic Stem Cells by Interleukin-27 into Myeloid Progenitors to Control Infection in Emergency Myelopoiesis

Jun-ichi Furusawa; Izuru Mizoguchi; Yukino Chiba; Masayuki Hisada; Fumie Kobayashi; Hiroki Yoshida; Susumu Nakae; Akihiko Tsuchida; Tetsuya Matsumoto; Hideo Ema; Junichiro Mizuguchi; Takayuki Yoshimoto

Emergency myelopoiesis is inflammation-induced hematopoiesis to replenish myeloid cells in the periphery, which is critical to control the infection with pathogens. Previously, pro-inflammatory cytokines such as interferon (IFN)-α and IFN-γ were demonstrated to play a critical role in the expansion of hematopoietic stem cells (HSCs) and myeloid progenitors, leading to production of mature myeloid cells, although their inhibitory effects on hematopoiesis were also reported. Therefore, the molecular mechanism of emergency myelopoiesis during infection remains incompletely understood. Here, we clarify that one of the interleukin (IL)-6/IL-12 family cytokines, IL-27, plays an important role in the emergency myelopoiesis. Among various types of hematopoietic cells in bone marrow, IL-27 predominantly and continuously promoted the expansion of only Lineage−Sca-1+c-Kit+ (LSK) cells, especially long-term repopulating HSCs and myeloid-restricted progenitor cells with long-term repopulating activity, and the differentiation into myeloid progenitors in synergy with stem cell factor. These progenitors expressed myeloid transcription factors such as Spi1, Gfi1, and Cebpa/b through activation of signal transducer and activator of transcription 1 and 3, and had enhanced potential to differentiate into migratory dendritic cells (DCs), neutrophils, and mast cells, and less so into macrophages, and basophils, but not into plasmacytoid DCs, conventional DCs, T cells, and B cells. Among various cytokines, IL-27 in synergy with the stem cell factor had the strongest ability to augment the expansion of LSK cells and their differentiation into myeloid progenitors retaining the LSK phenotype over a long period of time. The experiments using mice deficient for one of IL-27 receptor subunits, WSX-1, and IFN-γ revealed that the blood stage of malaria infection enhanced IL-27 expression through IFN-γ production, and the IL-27 then promoted the expansion of LSK cells, differentiating and mobilizing them into spleen, resulting in enhanced production of neutrophils to control the infection. Thus, IL-27 is one of the limited unique cytokines directly acting on HSCs to promote differentiation into myeloid progenitors during emergency myelopoiesis.


Cancer Science | 2015

Potential clinical application of interleukin-27 as an antitumor agent

Takayuki Yoshimoto; Yukino Chiba; Jun-ichi Furusawa; Mingli Xu; Ren Tsunoda; Kaname Higuchi; Izuru Mizoguchi

Cancer immunotherapies such as sipuleucel‐T and ipilimumab are promising new treatments that harness the power of the immune system to fight cancer and achieve long‐lasting remission. Interleukin (IL)‐27, a member of the IL‐12 heterodimeric cytokine family, has pleiotropic functions in the regulation of immune responses with both pro‐inflammatory and anti‐inflammatory properties. Evidence obtained using a variety of preclinical mouse models indicates that IL‐27 possesses potent antitumor activity against various types of tumors through multiple mechanisms without apparent adverse effects. These mechanisms include those mediated not only by CD8+ T cells, natural killer cells and macrophages, but also by antibody‐dependent cell‐mediated cytotoxicity, antiangiogenesis, direct antiproliferative effects, inhibition of expression of cyclooxygenase‐2 and prostaglandin E2, and suppression of epithelial–mesenchymal transition, depending on the characteristics of individual tumors. However, the endogenous role of IL‐27 subunits and one of its receptor subunits, WSX‐1, in the susceptibility to tumor development after transplantation of tumor cell lines or endogenously arising tumors seems to be more complicated. IL‐27 functions as a double‐edged sword: IL‐27 increases IL‐10 production and the expression of programmed death ligand 1 and T‐cell immunoglobulin and mucin domain‐3, and promotes the generation of regulatory T cells, and IL‐27 receptor α singling enhances transformation; IL‐27 may augment protumor effects as well. Here, we review both facets of IL‐27, antitumor effects and protumor effects, and discuss the potential clinical application of IL‐27 as an antitumor agent.


Immunobiology | 2013

IL-27 promotes nitric oxide production induced by LPS through STAT1, NF-κB and MAPKs

Motomu Shimizu; Kiyoshi Ogura; Izuru Mizoguchi; Yukino Chiba; Kaname Higuchi; Hiromi Ohtsuka; Junichiro Mizuguchi; Takayuki Yoshimoto

Interleukin (IL)-27, a member of the IL-6/IL-12 heterodimeric cytokine family, induces pro-inflammatory responses including early T helper (Th)1 differentiation and generation of cytotoxic T lymphocytes, and also anti-inflammatory responses including the differentiation to IL-10-producing regulatory T cells, inhibition of Th2 and Th17 differentiation, and suppression of pro-inflammatory cytokine production. Nitric oxide (NO) is a potent source of reactive nitrogen species that play an important role in killing intracellular pathogens and forms a crucial component of host defense. Inducible NO synthase (iNOS), which catalyzes the production of NO, is induced by a range of stimuli including cytokines and microbes. Recently, IL-27 was reported to play an anti-inflammatory role in microglia by blocking oncostatin M-induced iNOS expression and neuronal toxicity. In the present study, we investigated the effects of IL-27 on NO production in thioglycollate-elicited peritoneal macrophages. IL-27 together with lipopolysaccharide (LPS) induced morphological change into more spread and elongated cells and synergistically enhanced NO production. The combined stimulation also enhanced iNOS mRNA expression and the NO production was abrogated by an iNOS inhibitor, NG-monomethyl L-arginine. The synergistic NO production could be attributed to the augmented Toll-like receptor (TLR)4 mRNA expression by the combination. Signal transducer and activator of transcription (STAT)1 was indispensable for the morphological change and NO production. The combination induced nuclear factor κB (NF-κB) translocation into nuclear and phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK), and their inhibitors suppressed NO production. These results suggest that in contrast to the anti-proinflammatory role in microglia, IL-27 exerts a pro-inflammatory role by enhancing NO production in peritoneal macrophages stimulated with LPS through activation of STAT1, NF-κB and MAPKs.


PLOS ONE | 2013

IL-27 Enhances the Expression of TRAIL and TLR3 in Human Melanomas and Inhibits Their Tumor Growth in Cooperation with a TLR3 Agonist Poly(I:C) Partly in a TRAIL-Dependent Manner

Yukino Chiba; Izuru Mizoguchi; Kana Mitobe; Kaname Higuchi; Hiroshi Nagai; Chikako Nishigori; Junichiro Mizuguchi; Takayuki Yoshimoto

Interleukin (IL)-27 is a member of the IL-6/IL-12 cytokine family and possesses potent antitumor activity, which is mediated by multiple mechanisms. Toll-like receptor (TLR)3 is the critical sensor of the innate immune system that serves to identify viral double-stranded RNA. TLR3 is frequently expressed by various types of malignant cells, and recent studies reported that a synthetic TLR3 agonist, polyinosinic-polycytidylic acid [poly(I:C)], induces antitumor effects on malignant cells. In the present study, we have explored the effect of IL-27 on human melanomas and uncovered a previously unknown mechanism. We found that IL-27 inhibits in vitro tumor growth of human melanomas and greatly enhances the expression of TNF-related apoptosis inducing ligand (TRAIL) in a dose-dependent manner. Neutralizing antibody against TRAIL partly but significantly blocked the IL-27–mediated inhibition of tumor growth. In addition, IL-27 and poly(I:C) cooperatively augmented TRAIL expression and inhibited tumor growth. The cooperative effect could be ascribed to the augmented expression of TLR3, but not retinoic acid-inducible gene-I or anti-melanoma differentiation-associated gene 5, by IL-27. The inhibition of tumor growth by the combination was also significantly abrogated by anti-TRAIL neutralizing antibody. Moreover, IL-27 and poly(I:C) cooperatively suppressed in vivo tumor growth of human melanoma in immunodeficient mice. Taken together, these results suggest that IL-27 enhances the expression of TRAIL and TLR3 in human melanomas and inhibits their tumor growth in cooperation with poly(I:C), partly in a TRAIL-dependent manner. Thus, IL-27 and the combination of IL-27 and poly(I:C) may be attractive candidates for cancer immunotherapy.


Immunotherapy | 2009

Interleukins and cancer immunotherapy

Takayuki Yoshimoto; Noriko Morishima; Masae Okumura; Yukino Chiba; Mingli Xu; Junichiro Mizuguchi

Cancer is a complex disease with interactions between normal and neoplastic cells. Since current therapies for cancer largely rely on drugs or radiation that kill dividing cells or block cell division, these treatments may have severe side effects on normal proliferating cells in patients with cancer. Therefore, the potential for treatment of cancer patients by immunologic approaches, which may be specific for tumors and will not injure most normal cells, has great promise. Cancer immunotherapy aims to augment the weak host immune response to developing tumors. One strategy is to utilize cytokines such as IL-2. More recently, several exciting new interleukins have been characterized that have considerable promise for future immunotherapy. The promise of cancer immunotherapy largely depends upon the identification of these novel interleukins. This review provides an overview of the antitumor effects of relatively new interleukins as potential therapeutic agents applicable for cancer immunotherapy.

Collaboration


Dive into the Yukino Chiba's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mingli Xu

Tokyo Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kaname Higuchi

Tokyo Medical University

View shared research outputs
Top Co-Authors

Avatar

Mio Ohashi

Tokyo Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Toshiyuki Owaki

Tokyo University of Science

View shared research outputs
Researchain Logo
Decentralizing Knowledge