Network


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

Hotspot


Dive into the research topics where Ryuichi Amakawa is active.

Publication


Featured researches published by Ryuichi Amakawa.


Journal of Experimental Medicine | 2002

Interferon-α and Interleukin-12 Are Induced Differentially by Toll-like Receptor 7 Ligands in Human Blood Dendritic Cell Subsets

Tomoki Ito; Ryuichi Amakawa; Tsuneyasu Kaisho; Hiroaki Hemmi; Kenichirou Tajima; Kazutaka Uehira; Yoshio Ozaki; Hideyuki Tomizawa; Shizuo Akira; Shirou Fukuhara

Dendritic cells (DCs) play a crucial role in the immune responses against infections by sensing microbial invasion through toll-like receptors (TLRs). In humans, two distinct DC subsets, CD11c− plasmacytoid DCs (PDCs) and CD11c+ myeloid DCs (MDCs), have been identified and can respond to different TLR ligands, depending on the differential expression of cognate TLRs. In this study, we have examined the effect of TLR-7 ligands on human DC subsets. Both subsets expressed TLR-7 and could respond to TLR-7 ligands, which enhanced the survival of the subsets and upregulated the surface expression of costimulatory molecules such as CD40, CD80, and CD86. However, the cytokine induction pattern was distinct in that PDCs and MDCs produced interferon (IFN)-α and interleukin (IL)-12, respectively. In response to TLR-7 ligands, the Th1 cell supporting ability of both DC subsets was enhanced, depending on the cytokines the respective subsets produced. This study demonstrates that TLR-7 exerts its biological effect in a DC subset-specific manner.


Journal of Immunology | 2001

Differential Regulation of Human Blood Dendritic Cell Subsets by IFNs

Tomoki Ito; Ryuichi Amakawa; Muneo Inaba; Susumu Ikehara; Kayo Inaba; Shirou Fukuhara

Based on the relative expression of CD11c and CD1a, we previously identified subsets of dendritic cells (DCs) or DC precursors in human peripheral blood. A CD1a+/CD11c+ population (CD11c+ DCs), also called myeloid DCs, is an immediate precursor of Langerhans cells, whereas a CD1a−/CD11c− population (CD11c− DCs), sometimes called lymphoid DCs but better known as plasmacytoid DCs, is composed of type I IFN (IFN-αβ)-producing cells. Here, we investigate the effects of IFN-αβ and IFN-γ as well as other cytokines on CD11c+ and CD11c− DC subsets, directly isolated from the peripheral blood, instead of in vitro-generated DCs. IFN-γ and IFN-α, rather than GM-CSF, were the most potent cytokines for enhancing the maturation of CD11c+ DCs. Incubation of CD11c+ DCs with IFN-γ also resulted in increased IL-12 production, and this IL-12 allowed DCs to increase Th1 responses by alloreactive T cells. In contrast, IFN-α did not induce IL-12 but, rather, augmented IL-10 production. IFN-α-primed matured CD11c+ DCs induced IL-10-producing regulatory T cells; however, this process was independent of the DC-derived IL-10. On the other hand, IFN-α by itself neither matured CD11c− DCs nor altered the polarization of responding T cells, although this cytokine was a potent survival factor for CD11c− DCs. Unlike IFN-α, IL-3 was a potent survival factor and induced the maturation of CD11c− DCs. The IL-3-primed CD11c− DCs activated T cells to produce IL-10, IFN-γ, and IL-4. Thus, CD11c+ and CD11c− DC subsets play distinct roles in the cytokine network, especially their responses to IFNs.


Journal of Immunology | 2004

Plasmacytoid Dendritic Cells Regulate Th Cell Responses through OX40 Ligand and Type I IFNs

Tomoki Ito; Ryuichi Amakawa; Muneo Inaba; Toshiyuki Hori; Maiko Ota; Kengo Nakamura; Masashi Takebayashi; Michihiko Miyaji; Tomoo Yoshimura; Kayo Inaba; Shirou Fukuhara

Dendritic cells (DCs) show a functional plasticity in determining Th responses depending on their maturational stage or on maturational signals delivered to the DCs. Human plasmacytoid DCs (PDCs) can induce either Th1- or Th2-type immune responses upon exposure to viruses or IL-3, respectively. In this study we have investigated the Th-polarizing capacity of PDCs after short (24-h) or long (72-h) culture with stimuli and have assessed the expression and function of OX40 ligand (OX40L) in PDC-mediated Th polarization in addition to type I IFN-dependent responses. IL-3-treated PDCs expressed OX40L, but produced almost no IFN-α in response to T cell stimulation (CD40 ligand or T cell interaction), resulting in the preferential priming of Th2 cells through OX40L-dependent mechanisms. Meanwhile, PDCs were rapidly endowed by viral infection (Sendai virus) with a high potency to develop IFN-γ-producing Th cells depending on their capacity to residually produce IFN-α. Although Sendai virus-stimulated PDCs simultaneously expressed OX40L in their maturational process, the Th1-inducing effect of endogenous type I IFNs may overcome and thus conceal the OX40L-dependent Th2 responses. However, during maturation in response to Sendai virus over the longer 72-h period, the expression level of OX40L was up-regulated, whereas the residual IFN-α-producing ability was down-regulated, and consequently, the PDCs with prolonged Sendai virus stimulation induced Th2 responses to some extent. Thus, PDCs have the distinct means to dictate an appropriate response to environmental stimuli.


Immunology | 2003

Immunomodulatory effects of cyclosporin A on human peripheral blood dendritic cell subsets

Kenichirou Tajima; Ryuichi Amakawa; Tomoki Ito; Michihiko Miyaji; Masashi Takebayashi; Shirou Fukuhara

Cyclosporin A (CsA) is a potent immuno‐suppressant and is approved for the treatment of various disease conditions. The molecular biological mechanism of CsA has been investigated intensively in T cells and has been shown to involve the intracellular calcineurin pathway. Recently, it was reported that CsA has capacities to affect not only T cells but also antigen‐presenting cells such as B cells and dendritic cells (DCs). DCs are a master regulator of immune responses that have an integral capacity to prime naive T cells. In the present study, we investigated the biological effects of CsA on human peripheral blood DC subsets: CD11c+ myeloid and CD11c− lymphoid subsets. CsA inhibited the up‐regulation of co‐stimulatory molecules induced with or without microbial stimuli and CD40L on both CD11c+ and CD11c− subsets. In addition, CsA negatively regulated the endocytic activity of CD11c+ DC during the immature state. CsA inhibited the interleukin‐12 (IL‐12) production, but augmented the IL‐10 production from the LPS‐stimulated CD11c+ subset, whereas CsA reduced the interferon‐α (IFN‐α) production from the CD11c− subset infected with Sendai virus (SV). Both the LPS‐stimulated CD11c+ subset and SV‐infected CD11c− subset preferentially induced the development of IFN‐γ‐producing T helper‐type 1 (Th1) cells. Pretreatment of these DC subsets with CsA inhibited the Th1 skewing. These findings suggested a DC‐mediated mechanism of immunosupression by CsA.


Human Immunology | 2002

Roles of toll-like receptors in natural interferon-producing cells as sensors in immune surveillance

Tomoki Ito; Ryuichi Amakawa; Shirou Fukuhara

Natural IFN-alpha/beta producing cells (IPCs) play a central role in innate immunity against microbial infections. In primary immune responses, toll-like receptors (TLRs), as major pattern-recognition receptors, are essential for IPCs as well as other antigen presenting cell (APC) subsets to recognize microbes. IPCs unequivocally express TLR7 and TLR9, and can respond to the respective ligand to produce IFN-alpha/beta and to rapidly differentiate into dendritic cells (DCs). Thereby, IPCs can not only activate innate immune system but also provoke T cell responses. Thus, IPCs link innate and adaptive immunity through TLR system. In addition, recent work has revealed the regulatory system of DC subsets in response to microbial invasion. In this context, by the different but complementary expression profile of TLRs, IPCs together with myeloid APC subsets constitute a rational system of immune surveillance that can cover a wide variety of pathogens and enlarge immune adjuvant effects.


European Journal of Immunology | 2002

Lipid rafts as the signaling scaffold for NK cell activation: tyrosine phosphorylation and association of LAT with phosphatidylinositol 3-kinase and phospholipase C-γ following CD2 stimulation

Hiroshi Inoue; Michihiko Miyaji; Atsushi Kosugi; Masakazu Nagafuku; Toshiro Okazaki; Tsuneyo Mimori; Ryuichi Amakawa; Shirou Fukuhara; Naochika Domae; Eda T. Bloom; Hisanori Umehara

Natural killer (NK) cells participate in both innate and adaptive immunity through the prompt secretion of cytokines and ability to lyse virally infected cells or tumor cells. Although it has been well understood that lipid rafts (rafts) and a raft‐associated linker for activation of T cells (LAT) plays a central role in TCR signal transduction, there are still great gaps in our knowledge of the molecular events involved in NK cell activation. We show here that CD2 and rafts became polarized to the site of NK cell activation by CD2 cross‐linking or target cell binding using confocal microscopy, and LAT and a significant amount of CD2 colocalized in raft fractions of sucrose‐density gradient from an NK cell line, NK3.3. CD2 cross‐linking strongly induced tyrosine phosphorylation of LAT, resulting in increased association with phosphatidylinositol 3‐kinase (PI 3‐K) and phospholipase C‐γ1 (PLC‐γ1). In vitro binding studies using glutathione S‐transferase fusion proteins demonstrated that a large portion of the association between LAT and PI 3‐K or PLC‐γ1 was mediated through their SH2 domains in tyrosine phosphorylation‐dependent manner. Furthermore, disruption of lipid rafts by cholesterol depletion from cell membranes using methyl‐β‐cyclodextrin markedly reduced LAT tyrosine phosphorylation and NK cell functions, including cytotoxicity and granule exocytosis. These results document that modulation of raft integrity by aggregation of NK cell activating receptors, which leads to the formation of complexes of LAT with PI 3‐K and PLC‐γ1, is essential for the NK cell lytic mechanisms.


Arthritis & Rheumatism | 2001

Alteration of peripheral blood dendritic cells in patients with primary Sjögren's syndrome.

Yoshio Ozaki; Ryuichi Amakawa; Tomoki Ito; Hiroshi Iwai; Kenichirou Tajima; Kazutaka Uehira; Hideo Kagawa; Yoshiko Uemura; Toshio Yamashita; Shirou Fukuhara

OBJECTIVE We recently identified 3 fractions of human peripheral blood (PB) dendritic cells (DC), including the monocyte-associated fractions 1 and 2 (CD1a+,CD11c+ and CD1a-,CD11c+, respectively) and the lymphoid-associated fraction 3 (CD1a-,CD11c-). We attempted to determine whether these fractions were altered in Sjögrens syndrome (SS). METHODS We examined 23 patients with primary SS and 22 normal control subjects. DC were purified from PB and analyzed by flow cytometry. Immunohistochemical staining of labial salivary glands of SS patients was performed with monoclonal antibodies against fascin, which is known to be specific for DC. RESULTS The total numbers of PB DC and fraction 1 DC were decreased in SS. Immunohistochemical staining demonstrated that fascin+,CD11c+,HLA-DR+ mononuclear cells were present and scattered among numerous fascin-hyperfiltrating cells in SS patients. Interferon-gamma (IFNgamma)-producing Th1 cells were shown to be increased in both PB and salivary glands of patients, indicating the presence of general IFNgamma-producing Th1 polarization in SS. Furthermore, numbers of Thl cells were increased when naive T cells were cocultured with fraction 1 DC in vitro. CONCLUSION These findings suggest selective trafficking of fraction 1 DC into focal sites of inflammation and subsequent promotion of Th1 balance, suggesting a novel pathogenesis of SS.


Immunology | 2006

Prostaglandin E2 is a negative regulator on human plasmacytoid dendritic cells

Tomoki Ito; Yoshio Ozaki; Tsutomu Tanijiri; Takashi Yokoi; Kengo Nakamura; Masashi Takebayashi; Ryuichi Amakawa; Shirou Fukuhara

Prostaglandin E2 (PGE2), a major lipid derived from the metabolism of arachidonic acid, is an environmentally bioactive substance produced by inflammatory processes and acts as a cAMP up‐regulator that plays an important role in immune responses. It has been reported that PGE2 has the ability to inhibit the production of interleukin‐12 by myeloid dendritic cells (MDCs) and macrophages, and then induce preferential T helper type 2 (Th2) cell responses. However, little is known of the function of PGE2 for plasmacytoid dendritic cells (PDCs), which may contribute to the innate and adaptive immune response to viral infection, allergy and autoimmune diseases. In the present study, we compared the biological effect of PGE2 on human PDCs and MDCs. PGE2 caused the death of PDCs but MDCs survived. Furthermore, we found that, whereas PGE2 inhibited interferon‐α production by PDCs in response to virus or cytosine–phosphate–guanosine, it inhibited interelukin‐12 production by MDCs in response to lipopolysaccharide (LPS) or poly(I:C). Although both virus‐stimulated PDCs and LPS‐stimulated MDCs preferentially induced the development of interferon‐γ‐producing Th1 cells, pretreatment with PGE2 led both DC subsets to attenuate their Th1‐inducing capacity. These findings suggest that PGE2 represents a negative regulator on not only MDCs but also PDCs.


Journal of Leukocyte Biology | 2007

Hodgkin's Reed‐Sternberg cell line (KM‐H2) promotes a bidirectional differentiation of CD4+CD25+Foxp3+ T cells and CD4+ cytotoxic T lymphocytes from CD4+ naive T cells

Tsutomu Tanijiri; Toshiki Shimizu; Kazutaka Uehira; Takashi Yokoi; Hideki Amuro; Hiroyuki Sugimoto; Yoshitaro Torii; Kenichirou Tajima; Tomoki Ito; Ryuichi Amakawa; Shirou Fukuhara

A recent report revealed that a large population of Hodgkins lymphoma‐infiltrating lymphocytes (HLILs) consisted of regulatory T cells. In this study, we cocultured CD4+ naive T cells with KM‐H2, which was established as a Hodgkins Reed‐Sternberg cell line, to clarify their ability to induce CD25+Forkhead box P3+ (Foxp3+) T cells. The characteristic analyses of T cells cocultured with KM‐H2 revealed the presence of CD4+CD25+ T cells. They expressed CTLA‐4, glucocorticoid‐induced TNFR family‐related gene, and Foxp3 and could produce large amounts of IL‐10. Conversely, KM‐H2 also generated CD4+ CTLs, which expressed Granzyme B and T cell intracellular antigen‐1 in addition to Foxp3+ T cells. They exhibit a strong cytotoxic effect against the parental KM‐H2. In conclusion, KM‐H2 promotes a bidirectional differentiation of CD4+ naive T cells toward Foxp3+ T cells and CD4+ CTLs. In addition to KM‐H2, several cell lines that exhibit the APC function were able to generate Foxp3+ T cells and CD4+ CTLs. Conversely, the APC nonfunctioning cell lines examined did not induce both types of cells. Our findings suggest that the APC function of tumor cells is essential for the differentiation of CD4+ naive T cells into CD25+Foxp3+ T cells and CD4+ CTLs and at least partly explains the predominance of CD25+Foxp3+ T cells in HLILs and their contribution to a better prognosis. Therefore, in APC‐functioning tumors, including classical Hodgkin lymphomas, which generate Foxp3+ T cells and CD4+ CTLs, these T cell repertories play a beneficial role synergistically in disease stability.


Arthritis & Rheumatism | 2010

Statins, inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, function as inhibitors of cellular and molecular components involved in type I interferon production.

Hideki Amuro; Tomoki Ito; Rie Miyamoto; Hiroyuki Sugimoto; Yoshitaro Torii; Naoto Nakamichi; Chihiro Yamazaki; Katsuaki Hoshino; Tsuneyasu Kaisho; Yoshio Ozaki; Muneo Inaba; Ryuichi Amakawa; Shirou Fukuhara

OBJECTIVE Statins, which are used as cholesterol-lowering agents, have pleiotropic immunomodulatory properties. Although beneficial effects of statins have been reported in autoimmune diseases, the mechanisms of these immunomodulatory effects are still poorly understood. Type I interferons (IFNs) and plasmacytoid dendritic cells (PDCs) represent key molecular and cellular pathogenic components in autoimmune diseases such as systemic lupus erythematosus (SLE). Therefore, PDCs may be a specific target of statins in therapeutic strategies against SLE. This study was undertaken to investigate the immunomodulatory mechanisms of statins that target the IFN response in PDCs. METHODS We isolated human blood PDCs by flow cytometry and examined the effects of simvastatin and pitavastatin on PDC activation, IFNalpha production, and intracellular signaling. RESULTS Statins inhibited IFNalpha production profoundly and tumor necrosis factor alpha production modestly in human PDCs in response to Toll-like receptor ligands. The inhibitory effect on IFNalpha production was reversed by geranylgeranyl pyrophosphate and was mimicked by either geranylgeranyl transferase inhibitor or Rho kinase inhibitor, suggesting that statins exert their inhibitory actions through geranylgeranylated Rho inactivation. Statins inhibited the expression of phosphorylated p38 MAPK and Akt, and the inhibitory effect on the IFN response was through the prevention of nuclear translocation of IFN regulatory factor 7. In addition, statins had an inhibitory effect on both IFNalpha production by PDCs from SLE patients and SLE serum-induced IFNalpha production. CONCLUSION Our findings suggest a specific role of statins in controlling type I IFN production and a therapeutic potential in IFN-related autoimmune diseases such as SLE.

Collaboration


Dive into the Ryuichi Amakawa's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tomoki Ito

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar

Hitoshi Ohno

Takeda Pharmaceutical Company

View shared research outputs
Top Co-Authors

Avatar

Kazutaka Uehira

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yoshio Ozaki

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar

Hideki Amuro

Kansai Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rie Miyamoto

Kansai Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge