研究者業績

米山 光俊

ヨネヤマ ミツトシ  (Mitsutoshi Yoneyama)

基本情報

所属
千葉大学 真菌医学研究センター真菌症研究部門感染免疫分野 教授
学位
理学博士(1993年3月 大阪大学)

researchmap会員ID
0000035537

外部リンク

論文

 88
  • Keiko Shibata, Harune Moriizumi, Koji Onomoto, Yuka Kaneko, Takuya Miyakawa, Shuhei Zenno, Masaru Tanokura, Mitsutoshi Yoneyama, Tomoko Takahashi, Kumiko Ui-Tei
    Nucleic Acids Research 52(9) 5209-5225 2024年4月19日  査読有り
    Abstract RNA silencing is a post-transcriptional gene-silencing mechanism mediated by microRNAs (miRNAs). However, the regulatory mechanism of RNA silencing during viral infection is unclear. TAR RNA-binding protein (TRBP) is an enhancer of RNA silencing that induces miRNA maturation by interacting with the ribonuclease Dicer. TRBP interacts with a virus sensor protein, laboratory of genetics and physiology 2 (LGP2), in the early stage of viral infection of human cells. Next, it induces apoptosis by inhibiting the maturation of miRNAs, thereby upregulating the expression of apoptosis regulatory genes. In this study, we show that TRBP undergoes a functional conversion in the late stage of viral infection. Viral infection resulted in the activation of caspases that proteolytically processed TRBP into two fragments. The N-terminal fragment did not interact with Dicer but interacted with type I interferon (IFN) signaling modulators, such as protein kinase R (PKR) and LGP2, and induced ER stress. The end results were irreversible apoptosis and suppression of IFN signaling. Our results demonstrate that the processing of TRBP enhances apoptosis, reducing IFN signaling during viral infection.
  • Mitsutoshi Yoneyama, Hiroki Kato, Takashi Fujita
    Immunity 57(4) 731-751 2024年4月  査読有り招待有り
  • Im JH, Duic I, Yoshimizu SH, Onomoto K, Yoneyama M, Kato H, Fujita T
    Scientific Reports 13(1) 6318 2023年4月  査読有り
  • Kojima I, Onomoto K, Zuo WJ, Ozawa M, Okuya K, Naitou K, Izumi F, Okajima M, Fujiwara T, Ito N, Yoneyama M, Yamada K, Nishizono A, Sugiyama M, Fujita T, Masatani T
    Journal of Virology 96(18) e0081022 2022年9月  査読有り
    Rabies virus (RABV) is a neglected zoonotic pathogen that causes lethal infections in almost all mammalian hosts, including humans. Recently, RABV has been reported to induce intracellular formation of stress granules (SGs), also known as platforms that activate innate immune responses.
  • Mari T Iwasawa, Hideaki Miyachi, Seiichiro Wakabayashi, Takashi Sugihira, Reika Aoyama, Seitaro Nakagawa, Yuki Katayama, Mitsutoshi Yoneyama, Hiromitsu Hara, Yoichiro Iwakura, Masanori Matsumoto, Naohiro Inohara, Hanako Koguchi-Yoshioka, Manabu Fujimoto, Gabriel Núñez, Hiroyuki Matsue, Yuumi Nakamura, Shinobu Saijo
    International Immunology 34(8) 409-420 2022年5月31日  査読有り
    Abstract IL-17 plays important roles in host defense against Candida albicans at barrier surfaces and during invasive infection. However, the role of IL-17 in host defense after colonization of the epidermis, a main site of C. albicans infection remains poorly understood. Using a murine model of epicutaneous candidiasis without skin abrasion, we found that skin inflammation triggered by epidermal C. albicans colonization was self-limiting with fungal clearance completed by day 7 after inoculation in wild-type mice or animals deficient in IL-17A or IL-17F. In contrast, marked neutrophilic inflammation in the epidermis and impaired fungal clearance was observed in mice lacking both IL-17A and IL-17F. Clearance of C. albicans was independent of Dectin-1, Dectin-2, Card9, TLR2, and Myd88 in the epidermal colonization model. We found that group 3 innate lymphoid cells (ILC3s) and γδT cells were the major IL-17 producers in the epicutaneous candidiasis model. Analyses of Rag2−/− mice and Rag2−/−Il2rg−/− mice revealed that production of IL-17A and IL-17F by ILC3s was sufficient for C. albicans clearance. Finally, we found that depletion of neutrophils impaired C. albicans clearance in the epidermal colonization model. Taken together, these findings indicate a critical and redundant function of IL-17A and IL-17F produced by ILC3s in host defense against C. albicans in the epidermis. The results also suggest that epidermal C. albicans clearance is independent of innate immune receptors or that these receptors act redundantly in fungal recognition and clearance.
  • Onomoto K, Yoneyama M, Asakura S, Matsumoto S, Kaiho T
    Journal of Antibacterial and Antifungal Agents 50(3) 97-103 2022年3月  査読有り
  • Yushi Hayashi, Hidenori Suzuki, Wataru Nakajima, Ikuno Uehara, Atsuko Tanimura, Toshiki Himeda, Satoshi Koike, Tatsuya Katsuno, Shin-Ichiro Kitajiri, Naoto Koyanagi, Yasushi Kawaguchi, Koji Onomoto, Hiroki Kato, Mitsutoshi Yoneyama, Takashi Fujita, Nobuyuki Tanaka
    PLoS One 16(11) e026044318 2021年11月  査読有り
    Although sensorineural hearing loss (SHL) is relatively common, its cause has not been identified in most cases. Previous studies have suggested that viral infection is a major cause of SHL, especially sudden SHL, but the system that protects against pathogens in the inner ear, which is isolated by the blood-labyrinthine barrier, remains poorly understood. We recently showed that, as audiosensory receptor cells, cochlear hair cells (HCs) are protected by surrounding accessory supporting cells (SCs) and greater epithelial ridge (GER or Kölliker's organ) cells (GERCs) against viral infections. Here, we found that virus-infected SCs and GERCs induce HC death via production of the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL). Notably, the HCs expressed the TRAIL death receptors (DR) DR4 and DR5, and virus-induced HC death was suppressed by TRAIL-neutralizing antibodies. TRAIL-induced HC death was not caused by apoptosis, and was inhibited by necroptosis inhibitors. Moreover, corticosteroids, the only effective drug for SHL, inhibited the virus-induced transformation of SCs and GERCs into macrophage-like cells and HC death, while macrophage depletion also inhibited virus-induced HC death. These results reveal a novel mechanism underlying virus-induced HC death in the cochlear sensory epithelium and suggest a possible target for preventing virus-induced SHL.
  • Koji Onomoto, Kazuhide Onoguchi, Mitsutoshi Yoneyama
    Cellular & Molecular Immunology 18(3) 539-555 2021年1月18日  査読有り責任著者
    Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) are RNA sensor molecules that play essential roles in innate antiviral immunity. Among the three RLRs encoded by the human genome, RIG-I and melanoma differentiation-associated gene 5, which contain N-terminal caspase recruitment domains, are activated upon the detection of viral RNAs in the cytoplasm of virus-infected cells. Activated RLRs induce downstream signaling via their interactions with mitochondrial antiviral signaling proteins and activate the production of type I and III interferons and inflammatory cytokines. Recent studies have shown that RLR-mediated signaling is regulated by interactions with endogenous RNAs and host proteins, such as those involved in stress responses and posttranslational modifications. Since RLR-mediated cytokine production is also involved in the regulation of acquired immunity, the deregulation of RLR-mediated signaling is associated with autoimmune and autoinflammatory disorders. Moreover, RLR-mediated signaling might be involved in the aberrant cytokine production observed in coronavirus disease 2019. Since the discovery of RLRs in 2004, significant progress has been made in understanding the mechanisms underlying the activation and regulation of RLR-mediated signaling pathways. Here, we review the recent advances in the understanding of regulated RNA recognition and signal activation by RLRs, focusing on the interactions between various host and viral factors.
  • Yushi Hayashi, Hidenori Suzuki, Wataru Nakajima, Ikuno Uehara, Atsuko Tanimura, Toshiki Himeda, Satoshi Koike, Tatsuya Katsuno, Shin-Ichiro Kitajiri, Naoto Koyanagi, Yasushi Kawaguchi, Koji Onomoto, Hiroki Kato, Mitsutoshi Yoneyama, Takashi Fujita, Nobuyuki Tanaka
    Scientific reports 10(1) 6740-6740 2020年4月21日  査読有り
    To protect the audiosensory organ from tissue damage from the immune system, the inner ear is separated from the circulating immune system by the blood-labyrinth barrier, which was previously considered an immune-privileged site. Recent studies have shown that macrophages are distributed in the cochlea, especially in the spiral ligament, spiral ganglion, and stria vascularis; however, the direct pathogen defence mechanism used by audiosensory receptor hair cells (HCs) has remained obscure. Here, we show that HCs are protected from pathogens by surrounding accessory supporting cells (SCs) and greater epithelial ridge (GER or Kölliker's organ) cells (GERCs). In isolated murine cochlear sensory epithelium, we established Theiler's murine encephalomyelitis virus, which infected the SCs and GERCs, but very few HCs. The virus-infected SCs produced interferon (IFN)-α/β, and the viruses efficiently infected the HCs in the IFN-α/β receptor-null sensory epithelium. Interestingly, the virus-infected SCs and GERCs expressed macrophage marker proteins and were eliminated from the cell layer by cell detachment. Moreover, lipopolysaccharide induced phagocytosis of the SCs without cell detachment, and the SCs phagocytosed the bacteria. These results reveal that SCs function as macrophage-like cells, protect adjacent HCs from pathogens, and provide a novel anti-infection inner ear immune system.
  • Tomoko Takahashi, Yuko Nakano, Koji Onomoto, Mitsutoshi Yoneyama, Kumiko Ui-Tei
    Nucleic acids research 48(3) 1494-1507 2020年2月20日  査読有り
    During viral infection, viral nucleic acids are detected by virus sensor proteins including toll-like receptor 3 or retinoic acid-inducible gene I-like receptors (RLRs) in mammalian cells. Activation of these virus sensor proteins induces type-I interferon production and represses viral replication. Recently, we reported that an RLR family member, laboratory of genetics and physiology 2 (LGP2), modulates RNA silencing by interacting with an RNA silencing enhancer, TAR-RNA binding protein (TRBP). However, the biological implications remained unclear. Here, we show that LGP2 enhances apoptosis by upregulating apoptosis regulatory genes during viral infection. Sendai virus (SeV) infection increased LGP2 expression approximately 900 times compared to that in non-virus-infected cells. Then, the induced LGP2 interacted with TRBP, resulting in the inhibition of maturation of the TRBP-bound microRNA (miRNA) and its subsequent RNA silencing activity. Gene expression profiling revealed that apoptosis regulatory genes were upregulated during SeV infection: caspases-2, -8, -3 and -7, four cysteine proteases with key roles in apoptosis, were upregulated directly or indirectly through the repression of a typical TRBP-bound miRNA, miR-106b. Our findings may shed light on the mechanism of apoptosis, induced by the TRBP-bound miRNAs through the interaction of TRBP with LGP2, as an antiviral defense system in mammalian cells.
  • Kiyohiro Takahasi, Koji Onomoto, Masataka Horiuchi, Hiroki Kato, Takashi Fujita, Mitsutoshi Yoneyama
    Biochemical and biophysical research communications 517(4) 662-669 2019年10月1日  査読有り最終著者責任著者
    Upon viral infection, retinoic acid-inducible gene-I (RIG-I)-like receptors detect viral foreign RNAs and transmit anti-viral signals via direct interaction with the downstream mitochondrial adaptor molecule, interferon (IFN)-β promoter stimulator-1 (IPS-1), to inhibit viral replication. Although IPS-1 is known to form prion-like oligomers on mitochondria to activate signaling, the mechanisms that regulate oligomer formation remain unclear. Here, we identified an autoinhibitory domain (AD) at amino acids 180-349 to suppress oligomerization of IPS-1 in a resting state and regulate activation of downstream signaling. Size exclusion chromatography (SEC) analysis demonstrated that AD was required to suppress auto-oligomerization of the caspase recruitment domain (CARD) of IPS-1 via intramolecular interactions. This was supported by the observation that cleavage of a peptide bond between IPS-1 CARD and AD by Tobacco Etch virus (TEV) protease relieved autoinhibition. Conversely, deletion of this domain from IPS-1 enhanced signal activation in IFN-reporter assays, suggesting that IPS-1 AD played a critical role in the regulation of IPS-1-mediated anti-viral signal activation. These findings revealed novel molecular interactions involved in the tight regulation of innate anti-viral immunity.
  • Matsuo K, Haku A, Bi B, Takahashi H, Kamada N, Yaguchi T, Saijo S, Yoneyama M, Goto Y
    Microbiol Immunol 63(5) 155-163 2019年5月  査読有り
  • Takara Ohto, Manami Konishi, Hiroki Tanaka, Koji Onomoto, Mitsutoshi Yoneyama, Yuta Nakai, Kota Tange, Hiroki Yoshioka, Hidetaka Akita
    Biological & pharmaceutical bulletin 42(2) 299-302 2019年  査読有り
    While the use of in vitro-transcribed mRNA (IVT-mRNA) in therapeutics is a rapidly expanding area, the transfection of the exogenous IVT-mRNA is accompanied by a risk of immune activation. This immunological defense mechanism suppresses cellular translation process and can reduce transfection efficiency to a considerable extent. In the present study, we investigated the in vitro effects of Integrated Stress Response Inhibitor (ISRIB), and dexamethasone, a steroidal anti-inflammatory drug, on the transfection activity of a lipid nanoparticle (LNP) that was composed of ionizable lipids and IVT-mRNA. In the case of transfection to mouse embryonic fibroblast (MEF) cells, ISRIB mainly enhanced the transfection activity at an early stage of transfection (0-6 h). In contrast, dexamethasone caused an increase in transfection activity at intermediate-late stages of transfection (4-48 h). We also investigated the in vivo effects of dexamethasone using an LNP on that the IVT-mRNA and lipid-conjugated dexamethasone (Dex-Pal) were co-loaded. The intravenous administration of the LNP successfully enhanced the protein expression in a mouse liver by up to 6.6-fold. Collectively, the co-delivery of an anti-inflammatory drug is a promising approach for enhancing transfection efficiency of IVT-mRNA.
  • Tomoko Takahashi, Yuko Nakano, Koji Onomoto, Fuminori Murakami, Chiaki Komori, Yutaka Suzuki, Mitsutoshi Yoneyama, Kumiko Ui-Tei
    Nucleic acids research 46(17) 9134-9147 2018年9月28日  査読有り
    Here we show that laboratory of genetics and physiology 2 (LGP2) virus sensor protein regulates gene expression network of endogenous genes mediated by TAR-RNA binding protein (TRBP)-bound microRNAs (miRNAs). TRBP is an enhancer of RNA silencing, and functions to recruit precursor-miRNAs (pre-miRNAs) to Dicer that processes pre-miRNA into mature miRNA. Viral infection activates the antiviral innate immune response in mammalian cells. Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs), including RIG-I, melanoma-differentiation-associated gene 5 (MDA5), and LGP2, function as cytoplasmic virus sensor proteins during viral infection. RIG-I and MDA5 can distinguish between different types of RNA viruses to produce antiviral cytokines, including type I interferon. However, the role of LGP2 is controversial. We found that LGP2 bound to the double-stranded RNA binding sites of TRBP, resulting in inhibition of pre-miRNA binding and recruitment by TRBP. Furthermore, although it is unclear whether TRBP binds to specific pre-miRNA, we found that TRBP bound to particular pre-miRNAs with common structural characteristics. Thus, LGP2 represses specific miRNA activities by interacting with TRBP, resulting in selective regulation of target genes. Our findings show that a novel function of LGP2 is to modulate RNA silencing, indicating the crosstalk between RNA silencing and RLR signaling in mammalian cells.
  • Takahashi T, Nakano Y, Onomoto K, Yoneyama M, Ui-Tei K
    Genes 9(10) 511-511 2018年  査読有り
  • Mitsutoshi Yoneyama, Michihiko Jogi, Koji Onomoto
    JOURNAL OF BIOCHEMISTRY 159(3) 279-286 2016年3月  筆頭著者責任著者
    Activation of antiviral innate immunity is triggered by cellular pattern recognition receptors. Retinoic acid inducible gene-I (RIG-I)-like receptors (RLRs) detect viral non-self RNA in cytoplasm of virus-infected cells and play a critical role in the clearance of the invaded viruses through production of antiviral cytokines. Among the three known RLRs, RIG-I and melanoma differentiation-associated gene 5 recognize distinct non-self signatures of viral RNA and activate antiviral signaling. Recent reports have clearly described the molecular machinery underlying the activation of RLRs and interactions with the downstream adaptor, mitochondrial antiviral signaling protein (MAVS). RLRs and MAVS are thought to form large multimeric filaments around cytoplasmic organelles depending on the presence of Lys63-linked ubiquitin chains. Furthermore, RLRs have been shown to localize to stress-induced ribonucleoprotein aggregate known as stress granules and utilize them as a platform for recognition/activation of signaling. In this review, we will focus on the current understanding of RLR-mediated signal activation and the interactions with stress-induced RNA granules.
  • Oh, Seong-Wook, Onomoto, Koji, Wakimoto, Mai, Onoguchi, Kazuhide, Ishidate, Fumiyoshi, Fujiwara, Takahiro, Yoneyama, Mitsutoshi, Kato, Hiroki, Fujita, Takashi
    Plos Pathogens 12(2) 2016年  
  • T. Okazaki, M. Higuchi, K. Takeda, K. Iwatsuki-Horimoto, M. Kiso, M. Miyagishi, H. Yanai, A. Kato, M. Yoneyama, T. Fujita, T. Taniguchi, Y. Kawaoka, H. Ichijo, Y. Gotoh
    SCIENCE SIGNALING 8(388) ra78 2015年5月  査読有り
  • Mitsutoshi Yoneyama, Koji Onomoto, Michihiko Jogi, Teppei Akaboshi, Takashi Fujita
    CURRENT OPINION IN IMMUNOLOGY 32 48-53 2015年2月  査読有り筆頭著者責任著者
    In higher vertebrates, recognition of the non-self signature of invading viruses by genome-encoded pattern recognition receptors initiates antiviral innate immunity. Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) detect viral RNA as a non-self pattern in the cytoplasm and activate downstream signaling. Detection of viral RNA also activates stress responses resulting in stress granule-like aggregates, which facilitate RLR-mediated antiviral immunity. Among the three RLR family members RIG-I and melanoma differentiation-associated gene 5 (MDA5) recognize distinct viral RNA species with differential molecular machinery and activate signaling through mitochondrial antiviral signaling (MAVS, also known as IPS-1NISA/Cardif), which leads to the expression of cytokines including type I and Ill interferons (IFNs) to restrict viral propagation. In this review, we summarize receni knowledge regarding RNA recognition and signal transduction by RLRs and MAVS/IPS-1.
  • Okazaki, Tomohiko, Higuchi, Maiko, Takeda, Kohsuke, Iwatsuki-Horimoto, Kiyoko, Kiso, Maki, Miyagishi, Makoto, Yanai, Hideyuki, Kato, Atsushi, Yoneyama, Mitsutoshi, Fujita, Takashi, Taniguchi, Tadatsugu, Kawaoka, Yoshihiro, Ichijo, Hidenori, Gotoh, Yukiko
    Science Signaling 8(388) 2015年  
  • Ryo Narita, Kiyohiro Takahasi, Etsu Murakami, Emi Hirano, Seiji P. Yamamoto, Mitsutoshi Yoneyama, Hiroki Kato, Takashi Fujita
    PLOS PATHOGENS 10(10) e1004417 2014年10月  査読有り
    RIG-I-like receptor (RLR) plays a pivotal role in the detection of invading pathogens to initiate type I interferon (IFN) gene transcription. Since aberrant IFN production is harmful, RLR signaling is strictly regulated. However, the regulatory mechanisms are not fully understood. By expression cloning, we identified Pumilio proteins, PUM1 and PUM2, as candidate positive regulators of RIG-I signaling. Overexpression of Pumilio proteins and their knockdown augmented and diminished IFN-beta promoter activity induced by Newcastle disease virus (NDV), respectively. Both proteins showed a specific association with LGP2, but not with RIG-I or MDA5. Furthermore, all of these components were recruited to NDV-induced antiviral stress granules. Interestingly, biochemical analyses revealed that Pumilio increased double-stranded (ds) RNA binding affinity of LGP2; however, Pumilio was absent in the dsRNA-LGP2 complex, suggesting that Pumilio facilitates viral RNA recognition by LGP2 through its chaperon-like function. Collectively, our results demonstrate an unknown function of Pumilio in viral recognition by LGP2.
  • Akiko Yonekawa, Shinobu Saijo, Yoshihiko Hoshino, Yasunobu Miyake, Eri Ishikawa, Maho Suzukawa, Hiromasa Inoue, Masato Tanaka, Mitsutoshi Yoneyama, Masatsugu Oh-hora, Koichi Akashi, Sho Yamasaki
    IMMUNITY 41(3) 402-413 2014年9月  査読有り
    Mycobacteria possess various immunomodulatory molecules on the cell wall. Mannose-capped lipoarabinomannan (Man-LAM), amajor lipoglycan of Mycobacterium tuberculosis, has long been known to have both inhibitory and stimulatory effects on host immunity. However, the direct Man-LAM receptor that explains its pleiotropic activities has not been clearly identified. Here, we report that a C-type lectin receptor Dectin-2 (gene symbol Clec4n) is a direct receptor for Man-LAM. Man-LAM activated bonemarrow- derived dendritic cells (BMDCs) to produce pro-and anti-inflammatory cytokines, whereas it was completely abrogated in Clec4n(-/-) BMDCs. Man-LAM promoted antigen-specific T cell responses through Dectin-2 on DCs. Furthermore, Man-LAM induced experimental autoimmune encephalitis (EAE) as an adjuvant in mice, whereas Clec4n(-/-) mice were resistant. Upon mycobacterial infection, Clec4n(-/-) mice showed augmented lung pathology. These results demonstrate that Dectin-2 contributes to host immunity against mycobacterial infection through the recognition of Man-LAM.
  • Koji Onomoto, Mitsutoshi Yoneyama, Gabriel Fung, Hiroki Kato, Takashi Fujita
    TRENDS IN IMMUNOLOGY 35(9) 420-428 2014年9月  査読有り
    Viral infection triggers the activation of antiviral innate immune responses in mammalian cells. Viral RNA in the cytoplasm activates signaling pathways that result in the production of interferons (IFNs) and IFN-stimulated genes. Some viral infections have been shown to induce cytoplasmic granular aggregates similar to the dynamic ribonucleoprotein aggregates termed stress granules (SGs), suggesting that these viruses may utilize this stress response for their own benefit. By contrast, some viruses actively inhibit SG formation, suggesting an antiviral function for these structures. We review here the relationship between different viral infections and SG formation. We examine the evidence for antiviral functions for SGs and highlight important areas of inquiry towards understanding cellular stress responses to viral infection.
  • Ji-Seung Yoo, Kiyohiro Takahasi, Chen Seng Ng, Ryota Ouda, Koji Onomoto, Mitsutoshi Yoneyama, Janice Ching Lai, Simon Lattmann, Yoshikuni Nagamine, Tadashi Matsui, Kuniyoshi Iwabuchi, Hiroki Kato, Takashi Fujita
    PLOS PATHOGENS 10(3) e1004012 2014年3月  査読有り
    RIG-I is a DExD/H-box RNA helicase and functions as a critical cytoplasmic sensor for RNA viruses to initiate antiviral interferon (IFN) responses. Here we demonstrate that another DExD/H-box RNA helicase DHX36 is a key molecule for RIG-I signaling by regulating double-stranded RNA (dsRNA)-dependent protein kinase (PKR) activation, which has been shown to be essential for the formation of antiviral stress granule (avSG). We found that DHX36 and PKR form a complex in a dsRNA-dependent manner. By forming this complex, DHX36 facilitates dsRNA binding and phosphorylation of PKR through its ATPase/helicase activity. Using DHX36 KO-inducible MEF cells, we demonstrated that DHX36 deficient cells showed defect in IFN production and higher susceptibility in RNA virus infection, indicating the physiological importance of this complex in host defense. In summary, we identify a novel function of DHX36 as a critical regulator of PKR-dependent avSG to facilitate viral RNA recognition by RIG-I-like receptor (RLR).
  • Chen Seng Ng, Michihiko Jogi, Ji-Seung Yoo, Koji Onomoto, Satoshi Koike, Takuya Iwasaki, Mitsutoshi Yoneyama, Hiroki Kato, Takashi Fujita
    JOURNAL OF VIROLOGY 87(17) 9511-9522 2013年9月  査読有り
    In response to stress, cells induce ribonucleoprotein aggregates, termed stress granules (SGs). SGs are transient loci containing translation-stalled mRNA, which is eventually degraded or recycled for translation. Infection of some viruses, including influenza A virus with a deletion of nonstructural protein 1 (IAV Delta NS1), induces SG-like protein aggregates. Previously, we showed that IAV Delta NS1-induced SGs are required for efficient induction of type I interferon (IFN). Here, we investigated SG formation by different viruses using green fluorescent protein (GFP)-tagged Ras-Gap SH3 domain binding protein 1 (GFP-G3BP1) as an SG probe. HeLa cells stably expressing GFP-G3BP1 were infected with different viruses, and GFP fluorescence was monitored live with time-lapse microscopy. SG formations by different viruses was classified into 4 different patterns: no SG formation, stable SG formation, transient SG formation, and alternate SG formation. We focused on encephalomyocarditis virus (EMCV) infection, which exhibited transient SG formation. We found that EMCV disrupts SGs by cleavage of G3BP1 at late stages of infection (>8 h) through a mechanism similar to that used by poliovirus. Expression of a G3BP1 mutant that is resistant to the cleavage conferred persistent formation of SGs as well as an enhanced induction of IFN and other cytokines at late stages of infection. Additionally, knockdown of endogenous G3BP1 blocked SG formation with an attenuated induction of IFN and potentiated viral replication. Taken together, our findings suggest a critical role of SGs as an antiviral platform and shed light on one of the mechanisms by which a virus interferes with host stress and subsequent antiviral responses.
  • Miho Sekai, Shizue Tani-ichi, Mitsutoshi Yoneyama, Takashi Fujita, Tatsuo Kina, Koichi Ikuta
    Molecular Immunology 54(3-4) 378-385 2013年7月  査読有り
    The interaction between lymphocytes and stromal cells plays important roles in coordinated development of early lymphocytes. IL-7 is an essential cytokine for early lymphocyte development produced by stromal cells in the thymus and bone marrow. Although IL-7 is induced by interaction of early lymphocytes and stromal cells, its molecular basis is still unknown. To address this question, we employed co-culture system with an IL-7-dependent pre-B cell line, DW34, and a thymic stromal cell line, TSt-4. Co-culture with DW34 cells enhanced the levels of IL-7 transcripts in TSt-4 cells. Interestingly, the co-culture also induced transcripts of IFN-α and IFN-β but not of IFN-γ. In addition, exogenous IFN-β stimulation increased the levels of IL-7 transcripts in TSt-4 cells. Next, to elucidate the molecular mechanism of IL-7 induction, we analyzed the IL-7 promoter activity by reporter assay. The IL-7 promoter showed specific transcriptional activity in TSt-4 cells. An interferon-stimulated response element (ISRE) in the IL-7 promoter was essential for the induction of IL-7 transcription by both co-culture and IFN-β stimulation. Finally, overexpression of wild-type and dominant-negative forms of interferon regulatory factors (IRFs) activated and repressed, respectively, the IL-7 promoter in TSt-4 cells. Collectively, these results suggested that IRFs activated by lymphocyte adhesion induce IL-7 transcription through ISRE in stromal cells and that type I IFNs may be involved in the activation of IRFs. Thus, this study implied a physiological function of the IFN/IRF signal during lymphocyte development. © 2013.
  • Motoko Oarada, Takashi Miki, Shohei Kohno, Kanae Sakai, Takeshi Nikawa, Mitsutoshi Yoneyama, Tohru Gonoi
    JOURNAL OF NUTRITIONAL BIOCHEMISTRY 24(7) 1314-1323 2013年7月  査読有り
    Unhealthy eating behaviors increase the risk of metabolic diseases, but the underlying mechanisms are not fully elucidated. Because inflammation contributes to the pathogenesis of metabolic diseases, it is important to understand the effects of unhealthy eating on the inflammatory state. The objective of our present study was to address the effects of a fasting-refeeding regime, a model of irregular eating, on the hepatic inflammatory responses in mouse. The animals were fasted for 48 h and then refed either a standard or low-carbohydrate/high-fat diet. Inflammatory gene expression in the liver was then sequentially measured for the first 17 h after initiation of refeeding. To assess the roles of dietary carbohydrates and toll-like receptor 2 (TLR2) in the refeeding-induced inflammatory changes, gene expression levels in mice refed only carbohydrates (a-corn starch and sucrose) at different doses and in TLR2-deficient mice refed a standard diet were also analyzed. Refeeding with a standard diet increased the liver expression of Tlr2, proinflammatory mediators (Cxcl10, Cxcl1, Cxcl2, Icam-1) and negative regulators of TLR-signaling (A20 and Atf3). These increases were attenuated in mice refed a low-carbohydrate/high-fat diet. Refeeding only alpha-corn starch and sucrose also increased the expression of these inflammatory pathway genes depending on the doses. TLR2 deficiency significantly attenuated the refeeding-induced increase in the liver expression of Cxcl10, Cxcl1, Icam-1 and A20. These findings suggest that an irregular eating behavior can elicit a liver inflammatory response, which is at least partly mediated by TLR2, and that dietary carbohydrates play critical roles in this process. (C) 2013 Elsevier Inc. All rights reserved.
  • Yushi Hayashi, Koji Ondmoto, Ryo Narita, Mitsutoshi Yoneyama, Hiroki Kato, Takayuki Nakagawa, Juichi Ito, Akiko Taura, Takashi Fujita
    MICROBES AND INFECTION 15(8-9) 592-598 2013年7月  査読有り
    The inner ear has been regarded as an immunoprivileged site because of isolation by the blood-labyrinthine barrier. Several reports have indicated the existence of immune cells in the inner ear, but there are no reports showing immunocompetence of the cochlear tissue. In this report, we examined the potential involvement of retinoic acid inducible gene-I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5), which are critical for initiating antiviral innate immune responses. We found that RIG-I and MDA5 are expressed in the mouse cochlear sensory epithelium, including Hensen's and Claudius' cells. Ex vivo viral infection using Theiler's murine encephalomyelitis virus revealed that the virus replicates in these cells and that protein levels of RIG-I and MDA5 are up-regulated. Furthermore, the critical antiviral transcription factor, interferon (IFN) regulatory factor-3, is activated in the infected cells as judged by its nuclear translocation and the accumulation of type I IFN transcripts. These results strongly suggest that RIG-I and MDA5 participate in innate antiviral responses in cochlear tissue. (C) 2013 Institut Pasteur. Published by Elsevier Masson SAS. All rights reserved.
  • Shiori Takamatsu, Kazuhide Onoguchi, Koji Onomoto, Ryo Narita, Kiyohiro Takahasi, Fumiyoshi Ishidate, Takahiro K. Fujiwara, Mitsutoshi Yoneyama, Hiroki Kato, Takashi Fujita
    PLoS ONE 8(1) e53578 2013年1月15日  査読有り
    The innate immune system recognizes viral nucleic acids and stimulates cellular antiviral responses. Intracellular detection of viral RNA is mediated by the Retinoic acid inducible gene (RIG)-I Like Receptor (RLR), leading to production of type I interferon (IFN) and pro-inflammatory cytokines. Once cells are infected with a virus, RIG-I and MDA5 bind to viral RNA and undergo conformational change to transmit a signal through direct interaction with downstream CARD-containing adaptor protein, IFN-β promoter stimulator-1 (IPS-1, also referred as MAVS/VISA/Cardif). IPS-1 is composed of N-terminal Caspase Activation and Recruitment Domain (CARD), proline-rich domain, intermediate domain, and C-terminal transmembrane (TM) domain. The TM domain of IPS-1 anchors it to the mitochondrial outer membrane. It has been hypothesized that activated RLR triggers the accumulation of IPS-1, which forms oligomer as a scaffold for downstream signal proteins. However, the exact mechanisms of IPS-1-mediated signaling remain controversial. In this study, to reveal the details of IPS-1 signaling, we used an artificial oligomerization system to induce oligomerization of IPS-1 in cells. Artificial oligomerization of IPS-1 activated antiviral signaling without a viral infection. Using this system, we investigated the domain-requirement of IPS-1 for its signaling. We discovered that artificial oligomerization of IPS-1 could overcome the requirement of CARD and the TM domain. Moreover, from deletion- and point-mutant analyses, the C-terminal Tumor necrosis factor Receptor-Associated Factor (TRAF) binding motif of IPS-1 (aa. 453-460) present in the intermediate domain is critical for downstream signal transduction. Our results suggest that IPS-1 oligomerization is essential for the formation of a multiprotein signaling complex and enables downstream activation of transcription factors, Interferon Regulatory Factor 3 (IRF3) and Nuclear Factor-κB (NF-κB), leading to type I IFN and pro-inflammatory cytokine production. © 2013 Takamatsu et al.
  • Sayuri Nitta, Naoya Sakamoto, Mina Nakagawa, Sei Kakinuma, Kako Mishima, Akiko Kusano-Kitazume, Kei Kiyohashi, Miyako Murakawa, Yuki Nishimura-Sakurai, Seishin Azuma, Megumi Tasaka-Fujita, Yasuhiro Asahina, Mitsutoshi Yoneyama, Takashi Fujita, Mamoru Watanabe
    HEPATOLOGY 57(1) 46-58 2013年1月  査読有り
    Hepatitis C virus (HCV) infection blocks cellular interferon (IFN)-mediated antiviral signaling through cleavage of Cardif by HCV-NS3/4A serine protease. Like NS3/4A, NS4B protein strongly blocks IFN-beta production signaling mediated by retinoic acidinducible gene I (RIG-I); however, the underlying molecular mechanisms are not well understood. Recently, the stimulator of interferon genes (STING) was identified as an activator of RIG-I signaling. STING possesses a structural homology domain with flaviviral NS4B, which suggests a direct protein-protein interaction. In the present study, we investigated the molecular mechanisms by which NS4B targets RIG-Iinduced and STING-mediated IFN-beta production signaling. IFN-beta promoter reporter assay showed that IFN-beta promoter activation induced by RIG-I or Cardif was significantly suppressed by both NS4B and NS3/4A, whereas STING-induced IFN-beta activation was suppressed by NS4B but not by NS3/4A, suggesting that NS4B had a distinct point of interaction. Immunostaining showed that STING colocalized with NS4B in the endoplasmic reticulum. Immunoprecipitation and bimolecular fluorescence complementation (BiFC) assays demonstrated that NS4B specifically bound STING. Intriguingly, NS4B expression blocked the protein interaction between STING and Cardif, which is required for robust IFN-beta activation. NS4B truncation assays showed that its N terminus, containing the STING homology domain, was necessary for the suppression of IFN-beta promoter activation. NS4B suppressed residual IFN-beta activation by an NS3/4A-cleaved Cardif (Cardif1-508), suggesting that NS3/4A and NS4B may cooperate in the blockade of IFN-beta production. Conclusion: NS4B suppresses RIG-Imediated IFN-beta production signaling through a direct protein interaction with STING. Disruption of that interaction may restore cellular antiviral responses and may constitute a novel therapeutic strategy for the eradication of HCV. (HEPATOLOGY 2013;57:46-58)
  • Ouda, Ryota, Onomoto, Koji, Takahasi, Kiyohiro, Kato, Hiroki, Yoneyama, Mitstoshi, Fujita, Takashi
    Cytokine 63(3) 288-288 2013年  
  • Kazuaki Inoue, Kyoko Tsukiyama-Kohara, Chiho Matsuda, Mitsutoshi Yoneyama, Takashi Fujita, Shusuke Kuge, Makoto Yoshiba, Michinori Kohara
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 428(4) 494-499 2012年11月  査読有り
    Interferon regulatory factor-3 (IRF-3), a key transcriptional factor in the type I interferon system, is frequently impaired by hepatitis C virus (HCV), in order to establish persistent infection. However, the exact mechanism by which the virus establishes persistent infection has not been fully understood yet. The present study aimed to investigate the effects of various HCV proteins on IRF-3 activation, and elucidate the underlying mechanisms. To achieve this, full-length HCV and HCV subgenomic constructs corresponding to structural and each of the nonstructural proteins were transiently transfected into HepG2 cells. IFN-beta induction, plaque formation, and IRF-3 dimerization were elicited by Newcastle disease virus (NDV) infection. The expressions of IRF-3 homodimer and its monomer, Ser386-phosphorylated IRF-3, and HCV core protein were detected by immunofluorescence and western blotting. IFN-beta mRNA expression was quantified by real-time PCR (RT-PCR), and IRF-3 activity was measured by the levels of IRF-3 dimerization and phosphorylation, induced by NDV infection or polyriboinosinic:polyribocytidylic acid [poly(I:C)]. Switching of the expression of the complete HCV genome as well as the core proteins, El, E2, and NS2, suppressed IFN-beta mRNA levels and IRF-3 dimerization, induced by NDV infection. Our study revealed a crucial region of the HCV core protein, basic amino acid region 1 (BR1), to inhibit IRF-3 dimerization as well as its phosphorylation induced by NDV infection and poly (I:C), thus interfering with IRF-3 activation. Therefore, our study suggests that rescue of the IRF-3 pathway impairment may be an effective treatment for HCV infection. (C) 2012 Elsevier Inc. All rights reserved.
  • Fei Xing, Tomoh Matsumiya, Koji Onomoto, Ryo Hayakari, Tadaatsu Imaizumi, Hidemi Yoshida, Mitsutoshi Yoneyama, Takashi Fujita, Kei Satoh
    PLOS ONE 7(9) e45136 2012年9月  査読有り
    Mitochondrial antiviral signaling protein (MAVS) is an essential adaptor molecule that is responsible for antiviral signaling triggered by retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), leading to the induction of type I interferon in innate immunity. Previous studies have shown that certain viruses evade the innate immune response by cleaving the MAVS protein. However, little is known about how MAVS is regulated in response to foreign RNA, including both single-stranded (ss) and double-stranded (ds) RNA, because most previous reports have shown that the cleavage of MAVS is executed by proteases that are induced or activated by the invading RNA viruses. Here, we report that MAVS mRNA is degraded in response to polyinosinic-polycytidylic acid (polyI:C), a synthetic dsRNA, in A549 cells. RNA interference (RNAi) experiments revealed that both ssRNA- and dsRNA-associated pattern-recognition receptors (PRRs) were not involved in the degradation of MAVS mRNA. Foreign RNA also induced the transient degradation of the MAVS protein. In the resting state, the MAVS protein was protected from degradation by interferon regulatory factor 3 (IRF3); moreover, the dimerization of IRF3 appeared to be correlated with the rescue of protein degradation in response to polyI: C. The overexpression of MAVS enhanced interferon-beta (IFN-beta) expression in response to polyI: C, suggesting that the degradation of MAVS contributes to the suppression of the hyper-immune reaction in late-phase antiviral signaling. Taken together, these results suggest that the comprehensive regulation of MAVS in response to foreign RNA may be essential to antiviral host defenses.
  • Koji Onomoto, Michihiko Jogi, Ji-Seung Yoo, Ryo Narita, Shiho Morimoto, Azumi Takemura, Suryaprakash Sambhara, Atushi Kawaguchi, Suguru Osari, Kyosuke Nagata, Tomoh Matsumiya, Hideo Namiki, Mitsutoshi Yoneyama, Takashi Fujita
    PLOS ONE 7(8) e430331 2012年8月  査読有り責任著者
    Retinoic acid inducible gene I (RIG-I)-like receptors (RLRs) function as cytoplasmic sensors for viral RNA to initiate antiviral responses including type I interferon (IFN) production. It has been unclear how RIG-I encounters and senses viral RNA. To address this issue, we examined intracellular localization of RIG-I in response to viral infection using newly generated anti-RIG-I antibody. Immunohistochemical analysis revealed that RLRs localized in virus-induced granules containing stress granule (SG) markers together with viral RNA and antiviral proteins. Because of similarity in morphology and components, we termed these aggregates antiviral stress granules (avSGs). Influenza A virus (IAV) deficient in non-structural protein 1 (NS1) efficiently generated avSGs as well as IFN, however IAV encoding NS1 produced little. Inhibition of avSGs formation by removal of either the SG component or double-stranded RNA (dsRNA)-dependent protein kinase (PKR) resulted in diminished IFN production and concomitant enhancement of viral replication. Furthermore, we observed that transfection of dsRNA resulted in IFN production in an avSGs-dependent manner. These results strongly suggest that the avSG is the locus for non-self RNA sensing and the orchestration of multiple proteins is critical in the triggering of antiviral responses.
  • Maiko Kageyama, Kiyohiro Takahasi, Ryo Narita, Reiko Hirai, Mitsutoshi Yoneyama, Hiroki Kato, Takashi Fujita
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 415(1) 75-81 2011年11月  査読有り
    In virus-infected cells, viral RNA with non-self structural pattern is recognized by DExD/Hbox RNA helicase, RIG-I. Once RIG-I senses viral RNA, it triggers a signaling cascade, resulting in the activation of genes including type I interferon, which activates antiviral responses. Overexpression of N-terminal caspase activation and recruitment domain (CARD) is sufficient to activate signaling; however basal activity of full-length RIG-I is undetectable. The repressor domain (RD), initially identified as a.a. 735-925, is responsible for diminished basal activity; therefore, it is suggested that RIG-I is under auto-repression in uninfected cells and the repression is reversed upon its encounter with viral RNA. In this report, we further delimited RD to a.a. 747-801, which corresponds to a linker connecting the helicase and the C-terminal domain (CTD). Alanine substitutions of the conserved residues in the linker conferred constitutive activity to full-length RIG-I. We found that the constitutive active mutants do not exhibit ATPase activity, suggesting that ATPase is required for de-repression but not signaling itself. Furthermore, trypsin digestion of recombinant RIG-I revealed that the wild-type, but not linker mutant conforms to the trypsin-resistant structure, containing CARD and helicase domain. The result strongly suggests that the linker is responsible for maintaining RIG-I in a "closed" structure to minimize unwanted production of interferon in uninfected cells. These findings shed light on the structural regulation of RIG-I function. (C) 2011 Elsevier Inc. All rights reserved.
  • Ryota Ouda, Koji Onomoto, Kiyohiro Takahasi, Michael R. Edwards, Hiroki Kato, Mitsutoshi Yoneyama, Takashi Fujita
    JOURNAL OF BIOLOGICAL CHEMISTRY 286(29) 26210-U901 2011年7月  査読有り
    In mammals, viral infections are detected by innate immune receptors, including Toll-like receptor and retinoic acid inducible gene I (RIG-I)-like receptor (RLR), which activate the type I interferon (IFN) system. IFN essentially activates genes encoding antiviral proteins that inhibit various steps of viral replication as well as facilitate the subsequent activation of acquired immune responses. In this study, we investigated the expression of non-coding RNA upon viral infection or RLR activation. Using a microarray, we identified several microRNAs (miRNA) specifically induced to express by RLR signaling. As suggested by Bioinformatics (miRBase Target Data base), one of the RLR-inducible miRNAs, miR-23b, actually knocked down the expression of very low density lipoprotein receptor (VLDLR) and LDLR-related protein 5 (LRP5). Transfection of miR-23b specifically inhibited infection of rhinovirus 1B (RV1B), which utilizes the low density lipoprotein receptor (LDLR) family for viral entry. Conversely, introduction of anti-miRNA-23b enhanced the viral yield. Knockdown experiments using small interfering RNA (siRNA) revealed that VLDLR, but not LRP5, is critical for an efficient infection by RV1B. Furthermore, experiments with the transfection of infectious viral RNA revealed that miR-23b did not affect post-entry viral replication. Our results strongly suggest that RIG-I signaling results in the inhibitions of infections of RV1B through the miR-23b-mediated down-regulation of its receptor VLDLR.
  • Kazuhide Onoguchi, Mitsutoshi Yoneyama, Takashi Fujita
    JOURNAL OF INTERFERON AND CYTOKINE RESEARCH 31(1) 27-31 2011年1月  査読有り
    Retinoic acid-inducible gene-I (RIG-I), melanoma differentiation-associated 5 (MDA5), and laboratory of genetics and physiology 2 (LGP2) form a family of DExD/H box RNA helicases. RIG-I-like receptors (RLRs) are expressed ubiquitously at low levels, and their expression is induced by treatment with type I interferon (IFN) or a viral infection. RLRs function as sensors for the detection of viral RNA (such as double-stranded RNA) in the cytoplasm to initiate antiviral responses by producing type I and type III IFNs. Unlike Toll-like receptors, which sense exogenous pathogen-associated molecular patterns, RLRs detect cytoplasmic viral RNA. Because RLRs are IFN-inducible viral sensors, they are critical in amplifying antiviral responses.
  • Kiyohiro Takahasi, Masataka Horiuchi, Kiyonaga Fujii, Shingo Nakamura, Nobuo N. Noda, Mitsutoshi Yoneyama, Takashi Fujita, Fuyuhiko Inagaki
    GENES TO CELLS 15(8) 901-910 2010年8月  査読有り
    The transcription factor IRF-3 is activated by microbial invasions and produces a variety of cytokines including type-I interferon. Upon microbial infection, IRF-3 is phosphorylated at its C-terminal regulatory domain, then oligomerized, translocated into the nucleus, and here it binds to CBP/p300. Although a number of studies have been reported investigating the activation mechanism of IRF-3, there are a number of unresolved issues, especially on the phosphorylation sites, the oligomerization process and the binding mechanism with CBP/p300. In this report, the phosphorylated IRF-3 regulatory domain (IRF-3 RD) was prepared using the kinase IKK-i, and the active form of phosphorylated IRF-3 RD was identified. The paper also reports the crystal structure of the active form of the phosphorylated IRF-3 RD. Furthermore, the phosphorylation of Ser386 was found to be essential for its dimerization and binding with CBP/p300 using mutational analysis and mass spectrometry. Thus, we conclude that the phosphorylation of Ser386 is essential for activation of IRF-3.
  • Kazuhide Onoguchi, Koji Onomoto, Shiori Takamatsu, Michihiko Jogi, Azumi Takemura, Shiho Morimoto, Ilkka Julkunen, Hideo Namiki, Mitsutoshi Yoneyama, Takashi Fujita
    PLOS PATHOGENS 6(7) e1001012 2010年7月  査読有り
    In virus-infected cells, RIG-I-like receptor (RLR) recognizes cytoplasmic viral RNA and triggers innate immune responses including production of type I and III interferon (IFN) and the subsequent expression of IFN-inducible genes. Interferon-beta promoter stimulator 1 (IPS-1, also known as MAVS, VISA and Cardif) is a downstream molecule of RLR and is expressed on the outer membrane of mitochondria. While it is known that the location of IPS-1 is essential to its function, its underlying mechanism is unknown. Our aim in this study was to delineate the function of mitochondria so as to identify more precisely its role in innate immunity. In doing so we discovered that viral infection as well as transfection with 5'ppp-RNA resulted in the redistribution of IPS-1 to form speckle-like aggregates in cells. We further found that Mitofusin 1 (MFN1), a key regulator of mitochondrial fusion and a protein associated with IPS-1 on the outer membrane of mitochondria, positively regulates RLR-mediated innate antiviral responses. Conversely, specific knockdown of MFN1 abrogates both the virus-induced redistribution of IPS-1 and IFN production. Our study suggests that mitochondria participate in the segregation of IPS-1 through their fusion processes.
  • Takashi Satoh, Hiroki Kato, Yutaro Kumagai, Mitsutoshi Yoneyama, Shintaro Sato, Kazufumi Matsushita, Tohru Tsujimura, Takashi Fujita, Shizuo Akira, Osamu Takeuchi
    PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 107(4) 1512-1517 2010年1月  査読有り
    RNA virus infection is recognized by retinoic acid-inducible gene (RIG)-I- like receptors (RLRs), RIG-I, and melanoma differentiation-associated gene 5 (MDA5) in the cytoplasm. RLRs are comprised of N-terminal caspase-recruitment domains (CARDs) and a DExD/H-box helicase domain. The third member of the RLR family, LGP2, lacks any CARDs and was originally identified as a negative regulator of RLR signaling. In the present study, we generated mice lacking LGP2 and found that LGP2 was required for RIG-I- and MDA5-mediated antiviral responses. In particular, LGP2 was essential for type I IFN production in response to picornaviridae infection. Overexpression of the CARDs from RIG-I and MDA5 in Lgp2(-/-) fibroblasts activated the IFN-beta promoter, suggesting that LGP2 acts upstream of RIG-I and MDA5. We further examined the role of the LGP2 helicase domain by generating mice harboring a point mutation of Lys-30 to Ala (Lgp2(K30A/K30A)) that abrogated the LGP2 ATPase activity. Lgp2(K30A/K30A) dendritic cells showed impaired IFN-beta productions in response to various RNA viruses to extents similar to those of Lgp2(-/-) cells. Lgp2(-/-) and Lgp2(K30A/K30A) mice were highly susceptible to encephalomyocarditis virus infection. Nevertheless, LGP2 and its ATPase activity were dispensable for the responses to synthetic RNA ligands for MDA5 and RIG-I. Taken together, the present data suggest that LGP2 facilitates viral RNA recognition by RIG-I and MDA5 through its ATPase domain.
  • Kazuhide Onoguchi, Kiyohiro Takahasi, Mitsutoshi Yoneyama, Takashi Fujita
    in Viruses and Interferon: Current Research, Ed. Karen Mossman, Caister Academic Press , 2010 2010年  査読有り
  • Mitsutoshi Yoneyama, Takashi Fujita
    REVIEWS IN MEDICAL VIROLOGY 20(1) 4-22 2010年1月  査読有り筆頭著者
    Viral infections are detected by sensor molecules, which initiate innate antiviral responses, including the activation of type I interferons (IFNs) and proinflammatory cytokines. These cytokines are responsible for not only inhibiting viral replication in infected cells but also regulating the induction of adaptive immunity, leading to the swift eradication of viruses. Recent advances in the identification of pathogen receptors in the innate immune system have revealed that distinct types of sensors play a role in the detection of viral nucleic acids in different ways; Toll-like receptors (TLRs), which detect viral DNA or RNA in endosomal compartments in immune cells, retinoic acid inducible gene-l (RIG-D-like receptors (RLRs), which recognise viral RNA in the cytoplasm, and DNA sensors, which detect cytoplasmic viral DNA. Since these sensors have to exclusively recognise viral infections, it is intriguing to understand how they distinguish self nucleic acids from foreign viral ones. Here, we review the current knowledge of the recognition of viral nucleic acids by these sensor molecules and the signal transduction machinery. Copyright (C) 2009 John Wiley & Sons, Ltd.
  • Onoguchi K, Yoneyama M
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme 54(16 Suppl) 2226-2232 2009年12月  査読有り
  • Kiyohiro Takahasi, Hiroyuki Kumeta, Natsuko Tsuduki, Ryo Narita, Taeko Shigemoto, Reiko Hirai, Mitsutoshi Yoneyama, Masataka Horiuchi, Kenji Ogura, Takashi Fujita, Fuyuhiko Inagaki
    JOURNAL OF BIOLOGICAL CHEMISTRY 284(26) 17465-17474 2009年6月  査読有り
    The RIG-I like receptor (RLR) comprises three homologues: RIG-I (retinoic acid-inducible gene I), MDA5(melanoma differentiation-associated gene 5), and LGP2 (laboratory of genetics and physiology 2). Each RLR senses different viral infections by recognizing replicating viral RNA in the cytoplasm. The RLR contains a conserved C-terminal domain (CTD), which is responsible for the binding specificity to the viral RNAs, including double-stranded RNA (dsRNA) and 5'-triphosphated single-stranded RNA (5'ppp-ssRNA). Here, the solution structures of the MDA5 and LGP2 CTD domains were solved by NMR and compared with those of RIG-I CTD. The CTD domains each have a similar fold and a similar basic surface but there is the distinct structural feature of a RNA binding loop; The LGP2 and RIG-I CTD domains have a large basic surface, one bank of which is formed by the RNA binding loop. MDA5 also has a large basic surface that is extensively flat due to open conformation of the RNA binding loop. The NMR chemical shift perturbation study showed that dsRNA and 5'ppp-ssRNA are bound to the basic surface of LGP2 CTD, whereas dsRNA is bound to the basic surface of MDA5 CTD but much more weakly, indicating that the conformation of the RNA binding loop is responsible for the sensitivity to dsRNA and 5'ppp-ssRNA. Mutation study of the basic surface and the RNA binding loop supports the conclusion from the structure studies. Thus, the CTD is responsible for the binding affinity to the viral RNAs.
  • Taeko Shigemoto, Maiko Kageyama, Reiko Hirai, JiPing Zheng, Mitsutoshi Yoneyama, Takashi Fujita
    JOURNAL OF BIOLOGICAL CHEMISTRY 284(20) 13348-13354 2009年5月  査読有り
    Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are essential for detecting viral RNA and triggering antiviral responses, including production of type I interferon. We analyzed the phenotype of non-synonymous mutants of human RIG-I and MDA5 reported in databases by functional complementation in cell cultures. Of seven missense mutations of RIG-I, S183I, which occurs within the second caspase recruitment domain repeat, inactivated this domain and conferred a dominant inhibitory function. Of 10 mutants of MDA5, two exhibited loss of function. A nonsense mutation, E627*, resulted in deletion of the C-terminal region and double-stranded RNA (dsRNA) binding activity. Another loss of function mutation, I923V, which occurs within the C-terminal domain, did not affect dsRNA binding activity, suggesting a novel and essential role for this residue in the signaling. Remarkably, these mutations are implicated in resistance to type I diabetes. However, the A946T mutation of MDA5, which has been implicated in type I diabetes by previous genetic analyses, affected neither dsRNA binding nor IFN gene activation. These results provide new insights into the structure-function relationship of RIG-I-like receptors as well as into human RIG-I-like receptor polymorphisms, antiviral innate immunity, and autoimmune diseases.
  • Mitsutoshi Yoneyama, Takashi Fujita
    IMMUNOLOGICAL REVIEWS 227 54-65 2009年1月  査読有り
    Viral infection is detected by cellular sensor molecules as foreign nucleic acids and initiates innate antiviral responses, including the activation of proinflammatory cytokines and type I interferon (IFN). Recent identification of cytoplasmic viral sensors, such as retinoic acid-inducible gene-I-like receptors (RLRs), highlights their significance in the induction of antiviral innate immunity. Moreover, it is intriguing to understand how they can discriminate endogenous RNA from foreign viral RNA and initiate signaling cascades leading to the induction of type I IFNs. This review focuses on the current understanding of the molecular machinery underlying RNA recognition and subsequent signal transduction by RLRs.
  • Seigyoku Go, Mitsutoshi Yoneyama, Takashi Fujita
    Uirusu 58(2) 97-103 2008年12月  
    The innate immunity plays a crucial role in initial response to viral infection. Retinoic acid inducible gene-I (RIG-I) detects diverse viral RNAs in host cell and triggers immune response, producing antiviral cytokine, namely type I interferon (IFN). Recently, we analyzed the substrate RNAs and identified RNA binding domain of RIG-I. Here we discuss structural mechanism of intracellular RNA sensing system by RIG-I and function of RIG-I family molecules in the antiviral innate immunity.
  • Mitsutoshi Yoneyama, Koji Onmoto, Takashi Fujita
    ADVANCED DRUG DELIVERY REVIEWS 60(7) 841-846 2008年4月  査読有り
    Non-self RNA appearing in a cell as a result of viral replication is detected by a cytoplasmic sensor called RIG-I-like receptor (RLR). RLR consists of RIG-I, MDA5, and LGP2, which are DExD/H helicases. Dornain structures of RLR for detecting non-self RNA and for relaying signals downstream have been elucidated. Different viruses produce structurally different RNA species and are sensed differentially by RLR molecules to initiate antiviral responses and subsequent antigen-specific adaptive immunity. (c) 2008 Elsevier B.V. All rights reserved.
  • Kiyohiro Takahasi, Mitsutoshi Yoneyama, Tatsuya Nishihori, Reiko Hirai, Hiroyuki Kumeta, Ryo Narita, Michael Gale, Fuyuhiko Inagaki, Takashi Fujita
    MOLECULAR CELL 29(4) 428-440 2008年2月  査読有り
    A DExD/H protein, RIG-1, is critical in innate antiviral responses by sensing viral RNA. Here we show that RIG-I recognizes two distinct viral RNA patterns: double-stranded (ds) and 5'ppp single-stranded (ss) RNA. The binding of RIG-I with dsRNA or 5'ppp ssRNA in the presence of ATP produces a common structure, as suggested by protease digestion. Further analyses demonstrated that the C-terminal domain of RIG-I (CTD) recognizes these RNA patterns and CTD coincides with the autorepression domain. Structural analysis of CTD by NMR spectroscopy in conjunction with mutagenesis revealed that the basic surface of CTD with a characteristic cleft interacts with RIG-I ligands. Our results suggest that the bipartite structure of CTD regulates RIG-I on encountering viral RNA patterns.

MISC

 60

講演・口頭発表等

 43

共同研究・競争的資金等の研究課題

 17