研究者業績

尾野 本浩司

オノモト コウジ  (KOJI ONOMOTO)

基本情報

所属
千葉大学 真菌医学研究センター 助教
学位
理学博士(早稲田大学)

J-GLOBAL ID
201801011703354076
researchmap会員ID
B000322691

論文

 37
  • Keiko Shibata, Harune Moriizumi, Koji Onomoto, Yuka Kaneko, Takuya Miyakawa, Shuhei Zenno, Masaru Tanokura, Mitsutoshi Yoneyama, Tomoko Takahashi, Kumiko Ui-Tei
    Nucleic Acids Research 2024年4月19日  
    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.
  • 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
  • Isshu Kojima, Koji Onomoto, Wenjie Zuo, Makoto Ozawa, Kosuke Okuya, Kiyotada Naitou, Fumiki Izumi, Misuzu Okajima, Takuro Fujiwara, Naoto Ito, Mitsutoshi Yoneyama, Kentaro Yamada, Akira Nishizono, Makoto Sugiyama, Takashi Fujita, Tatsunori Masatani
    Journal of Virology 2022年9月7日  
    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.
  • 尾野本浩司, 米山光俊, 浅倉聡, 浅倉聡, 松本伸一, 海宝龍夫, 海宝龍夫
    日本防菌防黴学会誌 50(3) 2022年  
  • Koji Onomoto, Kazuhide Onoguchi, Mitsutoshi Yoneyama
    Cellular & molecular immunology 18(3) 539-555 2021年3月  
    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
    PloS one 16(11) e0260443 2021年  
    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.
  • 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  
    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  査読有り筆頭著者
    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.
  • 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.
  • Takahashi T, Nakano Y, Onomoto K, Yoneyama M, Ui-Tei K
    Genes 9(10) 511-511 2018年10月  査読有り
  • 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.
  • Mitsutoshi Yoneyama, Michihiko Jogi, Koji Onomoto
    JOURNAL OF BIOCHEMISTRY 159(3) 279-286 2016年3月  査読有り
    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.
  • Seong-Wook Oh, Koji Onomoto, Mai Wakimoto, Kazuhide Onoguchi, Fumiyoshi Ishidate, Takahiro Fujiwara, Mitsutoshi Yoneyama, Hiroki Kato, Takashi Fujita
    PLOS PATHOGENS 12(2) e1005444 2016年2月  査読有り
    RIG-I triggers antiviral responses by recognizing viral RNA (vRNA) in the cytoplasm. However, the spatio-temporal dynamics of vRNA sensing and signal transduction remain elusive. We investigated the time course of events in cells infected with Newcastle disease virus (NDV), a non-segmented negative-strand RNA virus. RIG-I was recruited to viral replication complexes (vRC) and triggered minimal primary type I interferon (IFN) production. RIG-I subsequently localized to antiviral stress granules (avSG) induced after vRC formation. The inhibition of avSG attenuated secondary IFN production, suggesting avSG as a platform for efficient vRNA detection. avSG selectively captured positive-strand vRNA, and poly(A)(+) RNA induced IFN production. Further investigations suggested that uncapped vRNA derived from read-through transcription was sensed by RIG-I in avSG. These results highlight how viral infections stimulate host stress responses, thereby selectively recruiting uncapped vRNA to avSG, in which RIG-I and other components cooperate in an efficient antiviral program.
  • Sayuki Iijima, Kentaro Matsuura, Tsunamasa Watanabe, Koji Onomoto, Takashi Fujita, Kyoko Ito, Etsuko Iio, Tomokatsu Miyaki, Kei Fujiwara, Noboru Shinkai, Atsunori Kusakabe, Mio Endo, Shunsuke Nojiri, Takashi Joh, Yasuhito Tanaka
    PLOS ONE 10(2) e0118000 2015年2月  査読有り
    The levels of expression of interferon-stimulated genes (ISGs) in liver are associated with response to treatment with pegylated interferon (PEG-IFN) plus ribavirin (RBV). However, associations between the responses of ISGs to IFN-based therapy and treatment efficacy or interleukin-28B (IL28B) genotype have not yet been determined. Therefore, we investigated the early responses of ISGs and interferon-lambdas (IFN-lambda s) in peripheral blood mononuclear cells (PBMCs) during PEG-IFN/RBV plus NS3/4 protease inhibitor (PI) therapy. We prospectively enrolled 50 chronic hepatitis C patients with HCV genotype 1, and collected PBMCs at baseline, 8 and 24 h after the initial administration of PEG-IFN/RBV/PI. Levels of mRNAs for selected ISGs and IFN-lambda s were evaluated by real-time PCR. All 31 patients with a favorable IL28B genotype and 13 of 19 with an unfavorable genotype achieved sustained virological responses (SVR). Levels of mRNA for A20, SOCS1, and SOCS3, known to suppress antiviral activity by interfering with the IFN signaling pathway, as well as IRF1 were significantly higher at 8 h in patients with an unfavorable IL28B genotype than in those with a favorable one (P = 0.007, 0.026, 0.0004, 0.0006, respectively), especially in the non-SVR group. Particularly, the fold-change of IRF1 at 8 h relative to baseline was significantly higher in non-SVR than in SVR cases with an unfavorable IL28B genotype (P = 0.035). In conclusion, levels of several mRNAs of genes suppressing antiviral activity in PBMCs during PEG-IFN/RBV/PI differed according to IL28B genotypes, paralleling treatment efficacy.
  • 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
    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.
  • 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.
  • 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.
  • Shinsuke Marumoto, Seiji P. Yamamoto, Hiroshi Nishimura, Koji Onomoto, Mitsuyoshi Yatagai, Kazufumi Yazaki, Takashi Fujita, Takashi Watanabe
    JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 60(36) 9106-9111 2012年9月  査読有り
    The germicidal activity of pyroligneous acid (PA) against a picornavirus, encephalomyocarditis virus (EMCV), was analyzed, and the component responsible for its disinfectant activity was identified. Bamboo PA (BPA) inactivated EMCV, but neutralization of BPA abolished this activity. Using liquid-liquid phase extraction and silica gel column chromatography, the hydrophobic active fraction of BPA was separated and its 12 major components were identified. The active fraction was reconstructed by mixing synthetic chemicals at the determined concentrations, and a subtraction series of one chemical from the complete mixture was prepared. An in vitro virus assay demonstrated that phenol was the sole germicidal component, and acetic acid augmented the phenol's inactivating activity resulting in >5-log decrease in EMCV infectivity. Considering the low environmental risk of PA, these findings suggest that BPA is a potentially useful agent for preventing viral epidemics in agricultural and human environments.
  • 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) e43031 2012年8月  査読有り
    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.
  • 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.
  • Koji Onomoto, Shiho Morimoto, Takahisa Kawaguchi, Hidenori Toyoda, Masami Tanaka, Masahiko Kuroda, Kazuko Uno, Takashi Kumada, Fumihiko Matsuda, Kunitada Shimotohno, Takashi Fujita, Yoshiki Murakami
    PLOS ONE 6(5) e19799 2011年5月  査読有り
    Background: Despite being expensive, the standard combination of pegylated interferon (Peg-IFN)-alpha and ribavirin used to treat chronic hepatitis C (CH) results in a moderate clearance rate and a plethora of side effects. This makes it necessary to predict patient outcome so as to improve the accuracy of treatment. Although the antiviral mechanism of genetically altered IL28B is unknown, IL28B polymorphism is considered a good predictor of IFN combination treatment outcome. Methodology: Using microarray, we quantified the expression profile of 237 IFN related genes in 87 CH liver biopsy specimens to clarify the relationship between IFN pathway and viral elimination, and to predict patients' clinical outcome. In 72 out of 87 patients we also analyzed IL28B polymorphism (rs8099917). Principal Findings: Five IFN related-genes (IFI27, IFI 44, ISG15, MX1, and OAS1) had expression levels significantly higher in nonresponders (NR) than in normal liver (NL) and sustained virological responders (SVR); this high expression was also frequently seen in cases with the minor (TG or GG) IL28B genotype. The expression pattern of 31 IFN related-genes also differed significantly between NR and NL. We predicted drug response in NR with 86.1% accuracy by diagonal linear discriminant analysis (DLDA). Conclusion: IFN system dysregulation before treatment was associated with poor IFN therapy response. Determining IFN related-gene expression pattern based on patients' response to combination therapy, allowed us to predict drug response with high accuracy. This method can be applied to establishing novel antiviral therapies and strategies for patients using a more individual approach.
  • Koji Onomoto, Kazuhide Onoguchi, Kiyohiro Takahasi, Takashi Fujita
    JOURNAL OF INTERFERON AND CYTOKINE RESEARCH 30(12) 875-881 2010年12月  査読有り
    Type I interferon (IFN) is produced in a variety of tissues in the body in response to viral infections. Recent studies have revealed that cytoplasmic receptors for viral (nonself) RNA are responsible for triggering IFN production. Different viruses activate different sensors. Numerous signaling adaptors are reported to participate in the regulation of the IFN gene's activation. In this paper, the role of free polyubiquitine chains in the activation of retinoic acid inducible gene I (RIG-I)-like receptors and the involvement of mitochondria as a signaling platform in the modulation of RIG-I-like receptor signaling is reviewed.
  • 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.
  • Onomoto K, Y oneyama M, Fujita T
    14th International Congress of Immunology. Kobe, Japan, August 22-27, 2010 2010年  査読有り
  • 尾野本浩司, 藤田尚志
    蛋白質 核酸 酵素 54(8) 901-907 2009年6月10日  査読有り
  • Onomoto K, Fujita T
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme 54(8 Suppl) 901-907 2009年6月  査読有り
  • 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.
  • Takashi Fujita, Kazuhide Onoguchi, Koji Onomoto, Reiko Hirai, Mitsutoshi Yoneyama
    BIOCHIMIE 89(6-7) 754-760 2007年6月  査読有り
    TLRs detect several classes of virus-associated molecules, such as ssRNA, CpG-DNA and dsRNA, and transduce signals leading to the production of IFN. Recently discovered cytoplasmic RNA helicases, RIG-I and MDA5, selectively sense viral RNA species. Gene disruption studies revealed the critical but non-redundant function of RIG-I and MDA5 in host antiviral responses. (C) 2007 Elsevier Masson SAS. All rights reserved.
  • K. Onomoto, M. Yoneyama, T. Fujita
    Current Topics in Microbiology and Immunology 316 193-205 2007年  査読有り
    The recognition of viral nucleic acids with pattern recognition receptors (PRRs) is the first step in inducing the innate immune system. Type I interferons (IFNs), central mediators in antiviral innate immunity, along with other cytokines and chemokines, disrupt virus replication. Recent studies indicated at least two distinct pathways for the induction of type I IFN by viral infection. Toll-like receptors (TLRs) are extracellular or endosomal PRRs for microbial pathogens, whereas retinoic acid-inducible gene-I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are novel intracellular PRRs for the viral dsRNA. In this review, we describe the distinct mechanisms inducing type I IFNs through TLRs and RIG-I/MDA5 pathways. © 2007 Springer-Verlag.
  • 尾野本浩司, 米山光俊, 藤田尚志
    日本臨床 64(7) 1236-1243 2006年7月1日  査読有り
  • K. Onomoto, K. Onoguchi, M. Yoneyama, T. Fujita
    PROCEEDINGS OF THE 6TH INTERNATIONAL CYTOKINE CONFERENCE 64(7) 3-+ 2006年  査読有り
    The recognition of viral nucleic acids with pattern recognition receptors (PRRs) is the first step to induce the innate immune system. Type I interferons (IFNs), central mediators in the antiviral innate immunity, are responsible for the induction of cytokines and chemokines that disrupt virus replication. Recent studies indicated that at least two distinct pathways for the induction of type I IFN by viral infection. Toll-like receptors (TLRs) are extracellular or endosomal PRRs for microbial pathogens whereas retinoic acid inducible gene-I (RIG-1) and melanoma differentiation-associated gene 5 (MDA5) are novel intracellular PRRs for viral dsRNA. In this report we describe the distinct mechanisms inducing type I IFNs through TLRs and RIG-I/MDA5 pathways. Higher organisms including humans are equipped to combat viruses using two kinds of immune responses: innate and adaptive immunity. Unlike adaptive immunity, which is characterized by its specificity and memory, innate immunity is provoked early in infection and is critical as an initial response. The type I interferon (IFN) system plays a major role in antiviral innate immunity(1)). Upon viral infection, type I IFN is secreted in body fluid and expands IFN response signals, resulting in the activation of various enzymes that prevent viral replication. In addition to antiviral activity, type I IFN has been known to exert various biological effects such as cell cycle regulation, differentiation and immune modulation. Furthermore, innate immune responses lead to the activation of specific cells with antigen-presenting functions to facilitate the initiation of adaptive immunity. The triggering of the IFN system is the activation of IFN genes. Since the initial discovery of type I IFN, the activation mechanism of the type I IFN gene has been a major focus of biologists. Although several double-stranded (ds)RNA binding proteins such as PKR have been attributed to the detection of replicating viral RNA, gene knockout studies do not support its role. Recent functional analyses of TLR revealed that TLRs function as pathogen receptors including those of viral origin(2)). LR3 has been identified as a receptor for exogenous dsRNA: however TLR3-deficient cells activate type I IFN genes normally, suggesting the existence of other receptor(s). Our expression cloning identified an RNA helicase as an essential receptor for virus-derived dsRNA (Fig. 1)(3)). We describe in this article the recently identified function of the RIG-I family of RNA helicases in innate immune reactions to infecting viruses.
  • 尾野本浩司, 米山光俊, 藤田尚志
    実験医学増刊, 23: 3061-3066, 羊土社 2005年  査読有り

MISC

 28

書籍等出版物

 7

講演・口頭発表等

 6

共同研究・競争的資金等の研究課題

 8