201 results on '"Takayuki Murata"'
Search Results
2. Epstein-Barr virus lytic gene BNRF1 promotes B-cell lymphomagenesis via IFI27 upregulation.
- Author
-
Ken Sagou, Yoshitaka Sato, Yusuke Okuno, Takahiro Watanabe, Tomoki Inagaki, Yashiro Motooka, Shinya Toyokuni, Takayuki Murata, Hitoshi Kiyoi, and Hiroshi Kimura
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Epstein-Barr virus (EBV) is a ubiquitous human lymphotropic herpesvirus that is causally associated with several malignancies. In addition to latent factors, lytic replication contributes to cancer development. In this study, we examined whether the lytic gene BNRF1, which is conserved among gamma-herpesviruses, has an important role in lymphomagenesis. We found that lymphoblastoid cell lines (LCLs) established by BNRF1-knockout EBV exhibited remarkably lower pathogenicity in a mice xenograft model than LCLs produced by wild-type EBV (LCLs-WT). RNA-seq analyses revealed that BNRF1 elicited the expression of interferon-inducible protein 27 (IFI27), which promotes cell proliferation. IFI27 knockdown in LCLs-WT resulted in excessive production of reactive oxygen species, leading to cell death and significantly decreased their pathogenicity in vivo. We also confirmed that IFI27 was upregulated during primary infection in B-cells. Our findings revealed that BNRF1 promoted robust proliferation of the B-cells that were transformed by EBV latent infection via IFI27 upregulation both in vitro and in vivo.
- Published
- 2024
- Full Text
- View/download PDF
3. Epstein-Barr virus: the molecular virology and the associated diseases
- Author
-
Takayuki Murata
- Subjects
ebv ,lymphoma ,carcinoma ,infectious mononucleosis ,autoimmune diseases ,Medicine (General) ,R5-920 - Abstract
Ever since its discovery as the first human oncogenic virus, Epstein-Barr virus (EBV) has been the focus of many researchers and is one of the best-studied pathogens. EBV is a major causative agent of Burkitt lymphoma, Hodgkin lymphoma, post-transplantation lymphoproliferative disorder, NK/T cell lymphoma, chronic active EBV disease, nasopharyngeal carcinoma, gastric carcinoma, and infectious mononucleosis. Although a truly comprehensive understanding of the virus and the associated disorders remains elusive, major breakthroughs in molecular cloning and omics analyses are shedding new light on this important virus. For example, EBV is now implicated in autoimmune diseases and neurodegenerative disorders. This review provides an overview of the molecular biology of EBV, the research history, the associated disorders, and the epidemiology.
- Published
- 2023
- Full Text
- View/download PDF
4. Generation of Recombinant Authentic Live Attenuated Human Rotavirus Vaccine Strain RIX4414 (Rotarix®) from Cloned cDNAs Using Reverse Genetics
- Author
-
Saori Fukuda, Masanori Kugita, Kanako Kumamoto, Yuki Akari, Yuki Higashimoto, Shizuko Nagao, Takayuki Murata, Tetsushi Yoshikawa, Koki Taniguchi, and Satoshi Komoto
- Subjects
human rotavirus ,live attenuated rotavirus vaccine ,Rotarix® ,reverse genetics ,Microbiology ,QR1-502 - Abstract
The live attenuated human rotavirus vaccine strain RIX4414 (Rotarix®) is used worldwide to prevent severe rotavirus-induced diarrhea in infants. This strain was attenuated through the cell culture passaging of its predecessor, human strain 89-12, which resulted in multiple genomic mutations. However, the specific molecular reasons underlying its attenuation have remained elusive, primarily due to the absence of a suitable reverse genetics system enabling precise genetic manipulations. Therefore, we first completed the sequencing of its genome and then developed a reverse genetics system for the authentic RIX4414 virus. Our experimental results demonstrate that the rescued recombinant RIX4414 virus exhibits biological characteristics similar to those of the parental RIX4414 virus, both in vitro and in vivo. This novel reverse genetics system provides a powerful tool for investigating the molecular basis of RIX4414 attenuation and may facilitate the rational design of safer and more effective human rotavirus vaccines.
- Published
- 2024
- Full Text
- View/download PDF
5. Growth Transformation of B Cells by Epstein-Barr Virus Requires IMPDH2 Induction and Nucleolar Hypertrophy
- Author
-
Atsuko Sugimoto, Takahiro Watanabe, Kazuhiro Matsuoka, Yusuke Okuno, Yusuke Yanagi, Yohei Narita, Seiyo Mabuchi, Hiroyuki Nobusue, Eiji Sugihara, Masaya Hirayama, Tomihiko Ide, Takanori Onouchi, Yoshitaka Sato, Teru Kanda, Hideyuki Saya, Yasumasa Iwatani, Hiroshi Kimura, and Takayuki Murata
- Subjects
EBV ,IMPDH2 ,nucleolar hypertrophy ,growth transformation ,MPA ,MMF ,Microbiology ,QR1-502 - Abstract
ABSTRACT The in vitro growth transformation of primary B cells by Epstein-Barr virus (EBV) is the initial step in the development of posttransplant lymphoproliferative disorder (PTLD). We performed electron microscopic analysis and immunostaining of primary B cells infected with wild-type EBV. Interestingly, the nucleolar size was increased by two days after infection. A recent study found that nucleolar hypertrophy, which is caused by the induction of the IMPDH2 gene, is required for the efficient promotion of growth in cancers. In the present study, RNA-seq revealed that the IMPDH2 gene was significantly induced by EBV and that its level peaked at day 2. Even without EBV infection, the activation of primary B cells by the CD40 ligand and interleukin-4 increased IMPDH2 expression and nucleolar hypertrophy. Using EBNA2 or LMP1 knockout viruses, we found that EBNA2 and MYC, but not LMP1, induced the IMPDH2 gene during primary infections. IMPDH2 inhibition by mycophenolic acid (MPA) blocked the growth transformation of primary B cells by EBV, leading to smaller nucleoli, nuclei, and cells. Mycophenolate mofetil (MMF), which is a prodrug of MPA that is approved for use as an immunosuppressant, was tested in a mouse xenograft model. Oral MMF significantly improved the survival of mice and reduced splenomegaly. Taken together, these results indicate that EBV induces IMPDH2 expression through EBNA2-dependent and MYC-dependent mechanisms, leading to the hypertrophy of the nucleoli, nuclei, and cells as well as efficient cell proliferation. Our results provide basic evidence that IMPDH2 induction and nucleolar enlargement are crucial for B cell transformation by EBV. In addition, the use of MMF suppresses PTLD. IMPORTANCE EBV infections cause nucleolar enlargement via the induction of IMPDH2, which are essential for B cell growth transformation by EBV. Although the significance of IMPDH2 induction and nuclear hypertrophy in the tumorigenesis of glioblastoma has been reported, EBV infection brings about the change quickly by using its transcriptional cofactor, EBNA2, and MYC. Moreover, we present here, for the novel, basic evidence that an IMPDH2 inhibitor, namely, MPA or MMF, can be used for EBV-positive posttransplant lymphoproliferative disorder (PTLD).
- Published
- 2023
- Full Text
- View/download PDF
6. Tegument proteins of Epstein-Barr virus: Diverse functions, complex networks, and oncogenesis
- Author
-
Takayuki Murata
- Subjects
EBV ,Tegument ,Envelopment ,Egress ,Infectivity ,Oncogenesis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
The tegument is the structure between the envelope and nucleocapsid of herpesvirus particles. Viral (and cellular) proteins accumulate to create the layers of the tegument. Some Epstein-Barr virus (EBV) tegument proteins are conserved widely in Herpesviridae, but others are shared only by members of the gamma-herpesvirus subfamily. As the interface to envelope and nucleocapsid, the tegument functions in virion morphogenesis and budding of the nucleocapsid during progeny production. When a virus particle enters a cell, enzymes such as kinase and deubiquitinase, and transcriptional activators are released from the virion to promote virus infection. Moreover, some EBV tegument proteins are involved in oncogenesis. Here, we summarize the roles of EBV tegument proteins, in comparison to those of other herpesviruses.
- Published
- 2023
- Full Text
- View/download PDF
7. Genome-wide CRISPR screen for HSV-1 host factors reveals PAPSS1 contributes to heparan sulfate synthesis
- Author
-
Takeshi Suzuki, Yoshitaka Sato, Yusuke Okuno, Fumi Goshima, Tadahisa Mikami, Miki Umeda, Takayuki Murata, Takahiro Watanabe, Koichi Watashi, Takaji Wakita, Hiroshi Kitagawa, and Hiroshi Kimura
- Subjects
Biology (General) ,QH301-705.5 - Abstract
A genome-wide CRISPR screen for HSV-1 host factors using near-haploid HAP1 cells revealed PAPSS1 as an essential factor for heparan sulfate biosynthesis and HSV-1 infection, and identified several other host factors also involved in both processes.
- Published
- 2022
- Full Text
- View/download PDF
8. Epstein–Barr virus tegument protein BGLF2 in exosomes released from virus-producing cells facilitates de novo infection
- Author
-
Yoshitaka Sato, Masahiro Yaguchi, Yusuke Okuno, Hanako Ishimaru, Ken Sagou, Somi Ozaki, Takeshi Suzuki, Tomoki Inagaki, Miki Umeda, Takahiro Watanabe, Masahiro Fujimuro, Takayuki Murata, and Hiroshi Kimura
- Subjects
Epstein–Barr virus ,Exosome ,BGLF2 ,De novo infection ,Extraviral particle ,Medicine ,Cytology ,QH573-671 - Abstract
Abstract Background Viruses must adapt to the environment of their host cells to establish infection and persist. Diverse mammalian cells, including virus-infected cells, release extracellular vesicles such as exosomes containing proteins and miRNAs, and use these vesicles to mediate intercellular communication. However, the roles of exosomes in viral infection remain unclear. Results We screened viral proteins to identify those responsible for the exosome-mediated enhancement of Epstein–Barr virus (EBV) infection. We identified BGLF2 protein encapsulated in exosomes, which were released by EBV-infected cells. BGLF2 protein is a tegument protein that exists in the space between the envelope and nucleocapsid, and it is released into the cytoplasm shortly after infection. BGLF2 protein-containing exosomes enhanced viral gene expression and repressed innate immunity, thereby supporting the EBV infection. Conclusions The EBV tegument protein BGLF2 is encapsulated in exosomes and released by infected cells to facilitate the establishment of EBV infection. These findings suggest that tegument proteins support viral infection not only between the envelope and nucleocapsid, as well as in extraviral particles such as exosomes. Graphical abstract Video abstract
- Published
- 2022
- Full Text
- View/download PDF
9. Ubiquitination of SARS-CoV-2 NSP6 and ORF7a Facilitates NF-κB Activation
- Author
-
Hironori Nishitsuji, Satoko Iwahori, Mariko Ohmori, Kunitada Shimotohno, and Takayuki Murata
- Subjects
inflammation ,NF-κB ,SARS-CoV-2 ,Microbiology ,QR1-502 - Abstract
ABSTRACT Patients with severe coronavirus disease 2019 tend to have high levels of proinflammatory cytokines, which eventually lead to cytokine storm and the development of acute respiratory distress syndrome. However, the detailed molecular mechanisms of proinflammatory cytokine production remain unknown. Here, we screened severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genes and found that nonstructural protein 6 (NSP6) and open reading frame 7a (ORF7a) activated the NF-κB pathway. NSP6 and ORF7a interacted with transforming growth factor β-activated kinase 1 (TAK1), and knockout (KO) of TAK1 or NF-κB essential modulator (NEMO) abolished NF-κB activation by NSP6 and ORF7a. Interestingly, K61 of NSP6 was conjugated to K63-linked polyubiquitin chains by the E3 ubiquitin ligase tripartite motif-containing 13, and this polyubiquitination of NSP6 appeared crucial for recruitment of NEMO to the NSP6-TAK1 complex and NF-κB activation. On the other hand, ring finger protein 121 (RNF121) was required for the polyubiquitination of ORF7a. Knockdown of RNF121 significantly decreased ORF7a binding of TAK1 and NEMO, resulting in the suppression of NF-κB activation. Taken together, our results provide novel molecular insights into the pathogenesis of SARS-CoV-2 and the host immune response to SARS-CoV-2 infection. IMPORTANCE The detailed molecular basis of the induction of proinflammatory cytokines and chemokines by SARS-CoV-2 is unclear, although such induction is clearly related to the severity of COVID-19. Here, we show that SARS-CoV-2 NSP6 and ORF7a lead to NF-κB activation through associations with TAK1. K63-linked polyubiquitination of NSP6 and ORF7a by TRIM13 and RNF121, respectively, appears essential for NF-κB activation. These results suggest that inhibition of the NSP6 and ORF7a gene products may reduce the severity of COVID-19 symptoms by decreasing proinflammatory cytokine levels.
- Published
- 2022
- Full Text
- View/download PDF
10. Oncolytic activity of naturally attenuated herpes-simplex virus HF10 against an immunocompetent model of oral carcinoma
- Author
-
Gaku Takano, Shinichi Esaki, Fumi Goshima, Atsushi Enomoto, Yoshimi Hatano, Haruka Ozaki, Takahiro Watanabe, Yoshitaka Sato, Daisuke Kawakita, Shingo Murakami, Takayuki Murata, Yukihiro Nishiyama, Shinichi Iwasaki, and Hiroshi Kimura
- Subjects
oncolytic viotherapy ,herpes simplex virus ,HF10 ,tongue cancer ,murine oral carcinoma ,4-nitroquinoline 1-oxide ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Prognosis for advanced oral carcinoma remains poor. Oncolytic virotherapy uses replication-competent viruses to infect and kill only the tumor cells. However, it has been difficult to investigate the oncolytic activity of viruses against oral carcinomas in mouse models. This study established a mouse model of oral cancer and investigated the in vitro and in vivo anti-tumor effects of HF10, a highly attenuated, replication-competent herpes simplex virus (HSV)-1. Mouse tongue cancer was induced by injecting 4-nitroquinoline 1-oxide into the mouse tongue. The murine oral cancer cell line isolated from this tumor, named NMOC1, formed invasive carcinoma within a week when injected into mouse tongue. HF10 successfully infected, replicated, and spread in the cancer cells in vitro. HF10 was able to kill cancer cells isolated from human or mouse tongue tumor. HF10 injection into tongue carcinomas prolonged mouse survival without any side effects or weight loss. Intertumoral injection of GFP-expressing HF10 confirmed that viral spread was confined within the tumors. Immunohistochemical staining showed that HF10 induced infiltration of CD8-positive T cells around HSV-infected cells in the tumor mass, implying increased anti-tumor immunity. We successfully established an oral cancer cell line and showed that HF10 is a promising therapeutic agent for oral cancer.
- Published
- 2021
- Full Text
- View/download PDF
11. EBV Exploits RNA m6A Modification to Promote Cell Survival and Progeny Virus Production During Lytic Cycle
- Author
-
Yusuke Yanagi, Takahiro Watanabe, Yuya Hara, Yoshitaka Sato, Hiroshi Kimura, and Takayuki Murata
- Subjects
EBV ,lytic cycle ,m6A modification ,METTL3 ,3-deazaadenosine ,Microbiology ,QR1-502 - Abstract
N6-methyladenosine (m6A) mediates various biological processes by affecting RNA stability, splicing, and translational efficiency. The roles of m6A modification in Epstein-Barr virus (EBV) infection in the lytic phase are unclear. Here, knockout of the m6A methyltransferase, N6-methyladenosine methyltransferase-like 3 (METTL3), or inhibition of methylation by DAA or UZH1a decreased the expression of viral lytic proteins and reduced progeny virion production. Interestingly, cell growth and viability were decreased by induction of the lytic cycle in METTL3-knockout or inhibitor-treated cells. Apoptosis was induced in those conditions possibly because of a decreased level of the anti-apoptotic viral protein, BHRF1. Therefore, m6A shows potential as a target of lytic induction therapy for EBV-associated cancers, including Burkitt lymphoma.
- Published
- 2022
- Full Text
- View/download PDF
12. Deletion of Viral microRNAs in the Oncogenesis of Epstein–Barr Virus-Associated Lymphoma
- Author
-
Hiroshi Kimura, Yusuke Okuno, Yoshitaka Sato, Takahiro Watanabe, and Takayuki Murata
- Subjects
EBV ,microRNA ,BART ,lymphomagenesis ,CAEBV ,ENKTL ,Microbiology ,QR1-502 - Abstract
Epstein–Barr virus (EBV), which encodes >80 genes and nearly 50 non-coding RNAs, is a double-stranded DNA virus. EBV is associated with various types of lymphomas and lymphoproliferative disorders not only of B-cell but also T/NK-cell origin. However, the oncogenic mechanism remains poorly understood, including the EBV receptors expressed on T/NK cells, relationship of EBV with host genes, and epigenetic regulation of EBV and host genes. The roles of host and viral non-coding RNAs during tumorigenesis have been elucidated. EBV encodes at least 49 mature microRNAs (miRNAs), of which 44 are located in BamHI-A rightward transcripts (BARTs) region, and the remaining five are located in BamHI-H rightward fragment 1. BART miRNAs modulate cell differentiation, proliferation, apoptosis, and the cell cycle, and they are considered positive regulators of oncogenesis. We and others have recently reported that EBV-positive lymphomas frequently possess large deletions in BART miRNA clusters, suggesting that some viral miRNAs have suppressive effects on oncogenesis, and that deletion of these miRNAs may aid lymphoma formation.
- Published
- 2021
- Full Text
- View/download PDF
13. Shedding of Viable Virus in Asymptomatic SARS-CoV-2 Carriers
- Author
-
Takayuki Murata, Aki Sakurai, Masahiro Suzuki, Satoshi Komoto, Tomihiko Ide, Takuma Ishihara, and Yohei Doi
- Subjects
Microbiology ,QR1-502 - Abstract
A growing number of studies suggest the potential role of asymptomatic SARS-CoV-2 carriers as a major driver of the COVID-19 pandemic; however, virological assessment of asymptomatic infection has largely been limited to reverse transcription-PCR (RT-PCR), which can be persistently positive without necessarily indicating the presence of viable virus (e.g., replication-competent virus). Here, we evaluated the infectivity of asymptomatic SARS-CoV-2 carriers by detecting SARS-CoV-2-induced cytopathic effects on Vero cells using longitudinally obtained nasopharyngeal samples from asymptomatic carriers.
- Published
- 2021
- Full Text
- View/download PDF
14. Direct Evidence of Abortive Lytic Infection-Mediated Establishment of Epstein-Barr Virus Latency During B-Cell Infection
- Author
-
Tomoki Inagaki, Yoshitaka Sato, Jumpei Ito, Mitsuaki Takaki, Yusuke Okuno, Masahiro Yaguchi, H. M. Abdullah Al Masud, Takahiro Watanabe, Kei Sato, Shingo Iwami, Takayuki Murata, and Hiroshi Kimura
- Subjects
EBV ,pre-latent phase ,abortive lytic infection ,fate mapping ,neo virology ,Microbiology ,QR1-502 - Abstract
Viral infection induces dynamic changes in transcriptional profiles. Virus-induced and antiviral responses are intertwined during the infection. Epstein-Barr virus (EBV) is a human gammaherpesvirus that provides a model of herpesvirus latency. To measure the transcriptome changes during the establishment of EBV latency, we infected EBV-negative Akata cells with EBV-EGFP and performed transcriptome sequencing (RNA-seq) at 0, 2, 4, 7, 10, and 14 days after infection. We found transient downregulation of mitotic division-related genes, reflecting reprogramming of cell growth by EBV, and a burst of viral lytic gene expression in the early phase of infection. Experimental and mathematical investigations demonstrate that infectious virions were not produced in the pre-latent phase, suggesting the presence of an abortive lytic infection. Fate mapping using recombinant EBV provided direct evidence that the abortive lytic infection in the pre-latent phase converges to latent infection during EBV infection of B-cells, shedding light on novel roles of viral lytic gene(s) in establishing latency. Furthermore, we find that the BZLF1 protein, which is a key regulator of reactivation, was dispensable for abortive lytic infection in the pre-latent phase, suggesting the divergent regulation of viral gene expressions from a productive lytic infection.
- Published
- 2021
- Full Text
- View/download PDF
15. In Silico Analysis and Synthesis of Nafamostat Derivatives and Evaluation of Their Anti-SARS-CoV-2 Activity
- Author
-
Kazuhiro J. Fujimoto, Daniel C. F. Hobbs, Miki Umeda, Akihiro Nagata, Rie Yamaguchi, Yoshitaka Sato, Ayato Sato, Kohsuke Ohmatsu, Takashi Ooi, Takeshi Yanai, Hiroshi Kimura, and Takayuki Murata
- Subjects
COVID-19 ,anti-SARS-CoV-2 agent ,TMPRSS2 ,nafamostat ,camostat ,Microbiology ,QR1-502 - Abstract
Inhibition of transmembrane serine protease 2 (TMPRSS2) is expected to block the spike protein-mediated fusion of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Nafamostat, a potent TMPRSS2 inhibitor as well as a candidate for anti-SARS-CoV-2 drug, possesses the same acyl substructure as camostat, but is known to have a greater antiviral effect. A unique aspect of the molecular binding of nafamostat has been recently reported to be the formation of a covalent bond between its acyl substructure and Ser441 in TMPRSS2. In this study, we investigated crucial elements that cause the difference in anti-SARS-CoV-2 activity of nafamostat and camostat. In silico analysis showed that Asp435 significantly contributes to the binding of nafamostat and camostat to TMPRSS2, while Glu299 interacts strongly only with nafamostat. The estimated binding affinity for each compound with TMPRSS2 was actually consistent with the higher activity of nafamostat; however, the evaluation of the newly synthesized nafamostat derivatives revealed that the predicted binding affinity did not correlate with their anti-SARS-CoV-2 activity measured by the cytopathic effect (CPE) inhibition assay. It was further shown that the substitution of the ester bond with amide bond in nafamostat resulted in significantly weakened anti-SARS-CoV-2 activity. These results strongly indicate that the ease of covalent bond formation with Ser441 in TMPRSS2 possibly plays a major role in the anti-SARS-CoV-2 effect of nafamostat and its derivatives.
- Published
- 2022
- Full Text
- View/download PDF
16. Molecular Basis of Epstein–Barr Virus Latency Establishment and Lytic Reactivation
- Author
-
Takayuki Murata, Atsuko Sugimoto, Tomoki Inagaki, Yusuke Yanagi, Takahiro Watanabe, Yoshitaka Sato, and Hiroshi Kimura
- Subjects
EBV ,latency ,reactivation ,transcription ,epigenetics ,oncogenesis ,Microbiology ,QR1-502 - Abstract
Epstein–Barr virus (EBV) is a causative agent of infectious mononucleosis and several types of cancer. Like other herpesviruses, it establishes an asymptomatic, life-long latent infection, with occasional reactivation and shedding of progeny viruses. During latency, EBV expresses a small number of viral genes, and exists as an episome in the host–cell nucleus. Expression patterns of latency genes are dependent on the cell type, time after infection, and milieu of the cell (e.g., germinal center or peripheral blood). Upon lytic induction, expression of the viral immediate-early genes, BZLF1 and BRLF1, are induced, followed by early gene expression, viral DNA replication, late gene expression, and maturation and egress of progeny virions. Furthermore, EBV reactivation involves more than just progeny production. The EBV life cycle is regulated by signal transduction, transcription factors, promoter sequences, epigenetics, and the 3D structure of the genome. In this article, the molecular basis of EBV latency establishment and reactivation is summarized.
- Published
- 2021
- Full Text
- View/download PDF
17. Human Rotavirus Reverse Genetics Systems to Study Viral Replication and Pathogenesis
- Author
-
Satoshi Komoto, Saori Fukuda, Takayuki Murata, and Koki Taniguchi
- Subjects
human rotavirus ,reverse genetics ,rescue T7 plasmid ,helper expression plasmid ,11 plasmid-only system ,Microbiology ,QR1-502 - Abstract
Human rotaviruses (HuRVAs) are highly important causes of acute gastroenteritis in infants and young children worldwide. A lack of reliable and reproducible reverse genetics systems for HuRVAs has limited a proper understanding of HuRVA biology and also the rational design of live-attenuated vaccines. Since the development of the first reverse genetics system for RVAs (partially plasmid-based reverse genetics system) in 2006, there have been many efforts with the goal of generating infectious recombinant HuRVAs entirely from cloned cDNAs. However, the establishment of a HuRVA reverse genetics system was very challenging until 2019. This review article provides an overview of the historical background of the recent development of long-awaited HuRVA reverse genetics systems, beginning with the generation of recombinant human-simian reassortant RVAs with the aid of a helper virus in 2006 and the generation of recombinant animal (simian) RVAs in a helper virus-free manner in 2017, and culminating in the generation of recombinant HuRVAs entirely from plasmid cDNAs in 2019. Notably, the original HuRVA reverse genetics system has already been optimized to increase the efficiency of virus generation. Although the application of HuRVA reverse genetics systems has only just been initiated, these technologies will help to answer HuRVA research questions regarding viral replication and pathogenicity that could not be addressed before, and to develop next-generation vaccines and intestine-specific rotaviral vectors.
- Published
- 2021
- Full Text
- View/download PDF
18. The Epstein-Barr Virus BRRF1 Gene Is Dispensable for Viral Replication in HEK293 cells and Transformation
- Author
-
Masahiro Yoshida, Takahiro Watanabe, Yohei Narita, Yoshitaka Sato, Fumi Goshima, Hiroshi Kimura, and Takayuki Murata
- Subjects
Medicine ,Science - Abstract
Abstract The Epstein-Barr virus (EBV) is a gamma-herpesvirus associated with several malignancies. It establishes a latent infection in B lymphocytes and is occasionally reactivated to enter the lytic cycle. Here we examined the role of the EBV gene BRRF1, which is expressed in the lytic state. We first confirmed, using a DNA polymerase inhibitor, that the BRRF1 gene is expressed with early kinetics. A BRRF1-deficient recombinant virus was constructed using a bacterial artificial chromosome system. No obvious differences were observed between the wild-type, BRRF1-deficient mutant and the revertant virus in HEK293 cells in terms of viral lytic protein expression, viral DNA synthesis, progeny production, pre-latent abortive lytic gene expression and transformation of primary B cells. However, reporter assays indicated that BRRF1 may activate transcription in promoter- and cell type-dependent manners. Taken together, BRRF1 is dispensable for viral replication in HEK293 cells and transformation of B cells, but it may have effects on transcription.
- Published
- 2017
- Full Text
- View/download PDF
19. Full genome characterization of novel DS-1-like G9P[8] rotavirus strains that have emerged in Thailand.
- Author
-
Saori Fukuda, Ratana Tacharoenmuang, Ratigorn Guntapong, Sompong Upachai, Phakapun Singchai, Tomihiko Ide, Riona Hatazawa, Karun Sutthiwarakom, Santip Kongjorn, Napa Onvimala, Kriangsak Ruchusatsawast, Pimpa Rungnopakun, Jutarat Mekmallika, Yoshiki Kawamura, Kazushi Motomura, Masashi Tatsumi, Naokazu Takeda, Takayuki Murata, Tetsushi Yoshikawa, Ballang Uppapong, Koki Taniguchi, and Satoshi Komoto
- Subjects
Medicine ,Science - Abstract
The emergence and rapid spread of unusual DS-1-like intergenogroup reassortant rotaviruses having G1/3/8 genotypes have been recently reported from major parts of the world (Africa, Asia, Australia, Europe, and the Americas). During rotavirus surveillance in Thailand, three novel intergenogroup reassortant strains possessing the G9P[8] genotype (DBM2017-016, DBM2017-203, and DBM2018-291) were identified in three stool specimens from diarrheic children. In the present study, we determined and analyzed the full genomes of these three strains. On full-genomic analysis, all three strains were found to share a unique genotype constellation comprising both genogroup 1 and 2 genes: G9-P[8]-I2-R2-C2-M2-A2-N2-T2-E2-H2. Phylogenetic analysis demonstrated that each of the 11 genes of the three strains was closely related to that of emerging DS-1-like intergenogroup reassortant, human, and/or locally circulating human strains. Thus, the three strains were suggested to be multiple reassortants that had acquired the G9-VP7 genes from co-circulating Wa-like G9P[8] rotaviruses in the genetic background of DS-1-like intergenogroup reassortant (likely equine-like G3P[8]) strains. To our knowledge, this is the first description of emerging DS-1-like intergenogroup reassortant strains having the G9P[8] genotype. Our observations will add to the growing insights into the dynamic evolution of emerging DS-1-like intergenogroup reassortant rotaviruses through reassortment.
- Published
- 2020
- Full Text
- View/download PDF
20. Human Herpesvirus and the Immune Checkpoint PD-1/PD-L1 Pathway: Disorders and Strategies for Survival
- Author
-
Takayuki Murata
- Subjects
HSV ,VZV ,CMV ,EBV ,KSHV ,PD-1 ,Biology (General) ,QH301-705.5 - Abstract
The immune system has evolved as a complex and efficient means of coping with extrinsic materials, such as pathogens and toxins, as well as intrinsic abnormalities, such as cancers. Although rapid and timely activation of the immune system is obviously important, regulated downregulation of the system is almost as significant as activation to prevent runaway immunity, such as allergies and hypercytokinemia. Therefore, the immune checkpoint programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) pathway is beneficial for the host. On the other hand, pathogens have evolved to evade host immunity by taking advantage of the PD-1/PD-L1 pathway. This review is focused on human herpesviruses, such as herpes simplex virus (HSV), cytomegalovirus (CMV), and Epstein–Barr virus (EBV), which cause various types of disorders, and their relationships with the PD-1/PD-L1 pathway. Understanding such relationships will be useful for developing preventative and therapeutic methods for disorders caused by herpesviruses.
- Published
- 2021
- Full Text
- View/download PDF
21. Epstein-Barr virus genome packaging factors accumulate in BMRF1-cores within viral replication compartments.
- Author
-
Atsuko Sugimoto, Yoriko Yamashita, Teru Kanda, Takayuki Murata, and Tatsuya Tsurumi
- Subjects
Medicine ,Science - Abstract
Productive replication of Epstein-Barr virus (EBV) during the lytic cycle occurs in discrete sites within nuclei, termed replication compartments. We previously proposed that replication compartments consist of two subnuclear domains: "ongoing replication foci" and "BMRF1-cores". Viral genome replication takes place in ongoing replication foci, which are enriched with viral replication proteins, such as BALF5 and BALF2. Amplified DNA and BMRF1 protein accumulate in BMRF1-cores, which are surrounded by ongoing replication foci. We here determined the locations of procapsid and genome-packaging proteins of EBV via three-dimensional (3D) surface reconstruction and correlative fluorescence microscopy-electron microscopy (FM-EM). The results revealed that viral factors required for DNA packaging, such as BGLF1, BVRF1, and BFLF1 proteins, are located in the innermost subdomains of the BMRF1-cores. In contrast, capsid structural proteins, such as BBRF1, BORF1, BDLF1, and BVRF2, were found both outside and inside the BMRF1-cores. Based on these observations, we propose a model in which viral procapsids are assembled outside the BMRF1-cores and subsequently migrate therein, where viral DNA encapsidation occurs. To our knowledge, this is the first report describing capsid assembly sites in relation to EBV replication compartments.
- Published
- 2019
- Full Text
- View/download PDF
22. Regulation of Epstein-Barr Virus Life Cycle and Cell Proliferation by Histone H3K27 Methyltransferase EZH2 in Akata Cells
- Author
-
Takaya Ichikawa, Yusuke Okuno, Yoshitaka Sato, Fumi Goshima, Hironori Yoshiyama, Teru Kanda, Hiroshi Kimura, and Takayuki Murata
- Subjects
EBV ,EZH2 ,histone methylation ,Microbiology ,QR1-502 - Abstract
ABSTRACT Epigenetic modifications play a pivotal role in the expression of the genes of Epstein-Barr virus (EBV). We found that de novo EBV infection of primary B cells caused moderate induction of enhancer of zeste homolog 2 (EZH2), the major histone H3 lysine 27 (K27) methyltransferase. To investigate the role of EZH2, we knocked out the EZH2 gene in EBV-negative Akata cells by the CRISPR/Cas9 system and infected the cells with EBV, followed by selection of EBV-positive cells. During the latent state, growth of EZH2-knockout (KO) cells was significantly slower after infection compared to wild-type controls, despite similar levels of viral gene expression between cell lines. After induction of the lytic cycle by anti-IgG, KO of EZH2 caused notable induction of expression of both latent and lytic viral genes, as well as increases in both viral DNA replication and progeny production. These results demonstrate that EZH2 is crucial for the intricate epigenetic regulation of not only lytic but also latent gene expression in Akata cells. IMPORTANCE The life cycle of EBV is regulated by epigenetic modifications, such as CpG methylation and histone modifications. Here, we found that the expression of EZH2, which encodes a histone H3K27 methyltransferase, was induced by EBV infection; therefore, we generated EZH2-KO cells to investigate the role of EZH2 in EBV-infected Akata B cells. Disruption of EZH2 resulted in increased expression of EBV genes during the lytic phase and, therefore, efficient viral replication and progeny production. Our results shed light on the mechanisms underlying reactivation from an epigenetic point of view and further suggest a role for EZH2 as a form of innate immunity that restricts viral replication in infected cells.
- Published
- 2018
- Full Text
- View/download PDF
23. BGLF2 Increases Infectivity of Epstein-Barr Virus by Activating AP-1 upon De Novo Infection
- Author
-
Natsuno Konishi, Yohei Narita, Fumiya Hijioka, H. M. Abdullah Al Masud, Yoshitaka Sato, Hiroshi Kimura, and Takayuki Murata
- Subjects
AP-1 ,BGLF2 ,BRLF1 ,EBV ,cell signaling ,Microbiology ,QR1-502 - Abstract
ABSTRACT Epstein-Barr virus (EBV) is a human gammaherpesvirus that causes infectious mononucleosis and several malignancies, such as endemic Burkitt lymphoma and nasopharyngeal carcinoma. Herpesviruses carry genes that can modify cell functions, including transcription and ubiquitination, thereby facilitating viral growth and survival in infected cells. Using a reporter screening system, we revealed the involvement of several EBV gene products in such processes. Of these, BGLF2 activated the AP-1 signaling pathway through phosphorylation of p38 and c-Jun N-terminal kinase (JNK). Knockout of the BGLF2 gene did not affect viral gene expression and viral genome DNA replication, but resulted in marked reduction of progeny titer. We also found that the BGLF2 disruption resulted in significant loss of infectivity upon de novo infection. Interestingly, expression of a binding partner, BKRF4, repressed the activation of AP-1 by BGLF2. These results shed light on the physiological role of the tegument protein BGLF2. IMPORTANCE Epstein-Barr virus (EBV), an oncogenic gammaherpesvirus, carries ~80 genes. While several genes have been investigated extensively, most lytic genes remain largely unexplored. Therefore, we cloned 71 EBV lytic genes into an expression vector and used reporter assays to screen for factors that activate signal transduction pathways, viral and cellular promoters. BGLF2 activated the AP-1 signaling pathway, likely by interacting with p38 and c-Jun N-terminal kinase (JNK), and increased infectivity of the virus. We also revealed that BKRF4 can negatively regulate AP-1 activity. Therefore, it is suggested that EBV exploits and modifies the AP-1 signaling pathway for its replication and survival.
- Published
- 2018
- Full Text
- View/download PDF
24. Characterization of a Suppressive Cis-acting Element in the Epstein–Barr Virus LMP1 Promoter
- Author
-
Masahiro Yoshida, Takayuki Murata, Keiji Ashio, Yohei Narita, Takahiro Watanabe, H. M. Abdullah Al Masud, Yoshitaka Sato, Fumi Goshima, and Hiroshi Kimura
- Subjects
EBV ,LMP1 ,promoter ,transcription ,EBV-BAC ,Microbiology ,QR1-502 - Abstract
Latent membrane protein 1 (LMP1) is a major oncogene encoded by Epstein–Barr virus (EBV) and is essential for immortalization of B cells by the virus. Previous studies suggested that several transcription factors, such as PU.1, RBP-Jκ, NFκB, EBF1, AP-2 and STAT, are involved in LMP1 induction; however, the means by which the oncogene is negatively regulated remains unclear. Here, we introduced short mutations into the proximal LMP1 promoter that includes recognition sites for the E-box and Ikaros transcription factors in the context of EBV-bacterial artificial chromosome. Upon infection, the mutant exhibited increased LMP1 expression and EBV-mediated immortalization of B cells. However, single mutations of either the E-box or Ikaros sites had limited effects on LMP1 expression and transformation. Our results suggest that this region contains a suppressive cis-regulatory element, but other transcriptional repressors (apart from the E-box and Ikaros transcription factors) may remain to be discovered.
- Published
- 2017
- Full Text
- View/download PDF
25. Initial Characterization of the Epstein–Barr Virus BSRF1 Gene Product
- Author
-
Yusuke Yanagi, H. M. Abdullah Al Masud, Takahiro Watanabe, Yoshitaka Sato, Fumi Goshima, Hiroshi Kimura, and Takayuki Murata
- Subjects
EBV ,BSRF1 ,Golgi apparatus ,secondary envelopment ,Microbiology ,QR1-502 - Abstract
Epstein–Barr virus (EBV) is a ubiquitous virus that causes infectious mononucleosis and several types of cancer, such as Burkitt lymphoma, T/NK-cell lymphoma, and nasopharyngeal carcinoma. As a herpesvirus, it encodes more than 80 genes, many of which have not been characterized. EBV BamHI S rightward reading frame 1 (BSRF1) encodes a tegument protein that, unlike its homologs herpes simplex virus unique long 51 (UL51) and human cytomegalovirus UL71, has not been extensively investigated. To examine the role of BSRF1, we prepared knockout and revertant strains using the bacterial artificial chromosome system. Unexpectedly, the disruption of the gene had little or no effect on EBV lytic replication and the transformation of primary B cells. However, the knockdown of BSRF1 in B95-8 cells decreased progeny production. An immunofluorescence assay revealed that BSRF1 localized to the Golgi apparatus in the cytoplasm, as did its homologs. BSRF1 also associated with BamHI G leftward reading frame 3.5 (BGLF3.5), BamHI B rightward reading frame 2 (BBRF2), and BamHI A leftward reading frame 1 (BALF1), and BALF1 was incorporated into the tegument fraction with BSRF1. Taken together, our results indicate that BSRF1 plays a role in secondary envelopment or virion egress in the cytoplasm, as do its homolog genes.
- Published
- 2019
- Full Text
- View/download PDF
26. Epstein-Barr virus infection-induced inflammasome activation in human monocytes.
- Author
-
Yuka Torii, Jun-Ichi Kawada, Takayuki Murata, Hironori Yoshiyama, Hiroshi Kimura, and Yoshinori Ito
- Subjects
Medicine ,Science - Abstract
Inflammasomes are cytoplasmic sensors that regulate the activity of caspase-1 and the secretion of interleukin-1β (IL-1β) or interleukin-18 (IL-18) in response to foreign molecules, including viral pathogens. They are considered to be an important link between the innate and adaptive immune responses. However, the mechanism by which inflammasome activation occurs during primary Epstein-Barr virus (EBV) infection remains unknown. Human B lymphocytes and epithelial cells are major targets of EBV, although it can also infect a variety of other cell types. In this study, we found that EBV could infect primary human monocytes and the monocyte cell line, THP-1, inducing inflammasome activation. We incubated cell-free EBV with THP-1 cells or primary human monocytes, then confirmed EBV infection using confocal microscopy and flow cytometry. Lytic and latent EBV genes were detected by real-time RT-PCR in EBV-infected monocytes. EBV infection of THP-1 cells and primary human monocytes induced caspase-dependent IL-1β production, while EBV infection of B-cell or T-cell lines did not induce IL-1β production. To identify the sensor molecule responsible for inflammasome activation during EBV infection, we examined the mRNA and the protein levels of NLR family pyrin domain-containing 3 (NLRP3), absent in melanoma 2 (AIM2), and interferon-inducible protein 16 (IFI16). Increased AIM2 levels were observed in EBV-infected THP-1 cells and primary human monocytes, whereas levels of IFI16 and NLRP3 did not show remarkable change. Furthermore, knockdown of AIM2 by small interfering RNA attenuated caspase-1 activation. Taken together, our results suggest that EBV infection of human monocytes induces caspase-1-dependent IL-1β production, and that AIM2, acting as an inflammasome, is involved in this response.
- Published
- 2017
- Full Text
- View/download PDF
27. Heat shock protein 90 inhibitors repress latent membrane protein 1 (LMP1) expression and proliferation of Epstein-Barr virus-positive natural killer cell lymphoma.
- Author
-
Takayuki Murata, Seiko Iwata, Mohammed Nure Alam Siddiquey, Tetsuhiro Kanazawa, Fumi Goshima, Daisuke Kawashima, Hiroshi Kimura, and Tatsuya Tsurumi
- Subjects
Medicine ,Science - Abstract
Epstein-Barr virus (EBV) LMP1 is a major oncoprotein expressed in latent infection. It functions as a TNFR family member and constitutively activates cellular signals, such as NFκB, MAPK, JAK/STAT and AKT. We here screened small molecule inhibitors and isolated HSP90 inhibitors, Radicicol and 17-AAG, as candidates that suppress LMP1 expression and cell proliferation not only in EBV-positive SNK6 Natural Killer (NK) cell lymphoma cells, but also in B and T cells. Tumor formation in immuno-defficient NOD/Shi-scid/IL-2Rγ(null) (NOG) mice was also retarded. These results suggest that HSP90 inhibitors can be alternative treatments for patients with EBV-positive malignancies.
- Published
- 2013
- Full Text
- View/download PDF
28. Unexpected instability of family of repeats (FR), the critical cis-acting sequence required for EBV latent infection, in EBV-BAC systems.
- Author
-
Teru Kanda, Sachiko Shibata, Satoru Saito, Takayuki Murata, Hiroki Isomura, Hironori Yoshiyama, Kenzo Takada, and Tatsuya Tsurumi
- Subjects
Medicine ,Science - Abstract
A group of repetitive sequences, known as the Family of Repeats (FR), is a critical cis-acting sequence required for EBV latent infection. The FR sequences are heterogeneous among EBV strains, and they are sometimes subject to partial deletion when subcloned in E. coli-based cloning vectors. However, the FR stability in EBV-BAC (bacterial artificial chromosome) system has never been investigated. We found that the full length FR of the Akata strain EBV was not stably maintained in a BAC vector. By contrast, newly obtained BAC clones of the B95-8 strain of EBV stably maintained the full length FR during recombinant virus production and B-cell transformation. Investigation of primary DNA sequences of Akata-derived EBV-BAC clones indicates that the FR instability is most likely due to a putative secondary structure of the FR region. We conclude that the FR instability in EBV-BAC clones can be a pitfall in E. coli-mediated EBV genetics.
- Published
- 2011
- Full Text
- View/download PDF
29. The human cytomegalovirus UL76 gene regulates the level of expression of the UL77 gene.
- Author
-
Hiroki Isomura, Mark F Stinski, Takayuki Murata, Sanae Nakayama, Shigeki Chiba, Yoshiki Akatsuka, Teru Kanda, and Tatsuya Tsurumi
- Subjects
Medicine ,Science - Abstract
Human cytomegalovirus (HCMV) can be reactivated under immunosuppressive conditions causing several fatal pneumonitis, hepatitis, retinitis, and gastrointestinal diseases. HCMV also causes deafness and mental retardation in neonates when primary infection has occurred during pregnancy. In the genome of HCMV at least 194 known open reading frames (ORFs) have been predicted, and approximately one-quarter, or 41 ORFs, are required for viral replication in cell culture. In contrast, the majority of the predicted ORFs are nonessential for viral replication in cell culture. However, it is also possible that these ORFs are required for the efficient viral replication in the host. The UL77 gene of HCMV is essential for viral replication and has a role in viral DNA packaging. The function of the upstream UL76 gene in the HCMV-infected cells is not understood. UL76 and UL77 are cistons on the same viral mRNA and a conventional 5' mRNA for UL77 has not been detected. The vast majority of eukaryotic mRNAs are monocistronic, i.e., they encode only a single protein.To determine whether the UL76 ORF affects UL77 gene expression, we mutated UL76 by ORF frame-shifts, stop codons or deletion of the viral gene. The effect on UL77 protein expression was determined by either transfection of expression plasmids or infection with recombinant viruses. Mutation of UL76 ORF significantly increased the level of UL77 protein expression. However, deletion of UL76 upstream of the UL77 ORF had only marginal effects on viral growth.While UL76 is not essential for viral replication, the UL76 ORF is involved in regulation of the level of UL77 protein expression in a manner dependent on the translation re-initiation. UL76 may fine-tune the UL77 expression for the efficient viral replication in the HCMV- infected cells.
- Published
- 2010
- Full Text
- View/download PDF
30. Degradation of phosphorylated p53 by viral protein-ECS E3 ligase complex.
- Author
-
Yoshitaka Sato, Takumi Kamura, Noriko Shirata, Takayuki Murata, Ayumi Kudoh, Satoko Iwahori, Sanae Nakayama, Hiroki Isomura, Yukihiro Nishiyama, and Tatsuya Tsurumi
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
p53-signaling is modulated by viruses to establish a host cellular environment advantageous for their propagation. The Epstein-Barr virus (EBV) lytic program induces phosphorylation of p53, which prevents interaction with MDM2. Here, we show that induction of EBV lytic program leads to degradation of p53 via an ubiquitin-proteasome pathway independent of MDM2. The BZLF1 protein directly functions as an adaptor component of the ECS (Elongin B/C-Cul2/5-SOCS-box protein) ubiquitin ligase complex targeting p53 for degradation. Intringuingly, C-terminal phosphorylation of p53 resulting from activated DNA damage response by viral lytic replication enhances its binding to BZLF1 protein. Purified BZLF1 protein-associated ECS could be shown to catalyze ubiquitination of phospho-mimetic p53 more efficiently than the wild-type in vitro. The compensation of p53 at middle and late stages of the lytic infection inhibits viral DNA replication and production during lytic infection, suggesting that the degradation of p53 is required for efficient viral propagation. Taken together, these findings demonstrate a role for the BZLF1 protein-associated ECS ligase complex in regulation of p53 phosphorylated by activated DNA damage signaling during viral lytic infection.
- Published
- 2009
- Full Text
- View/download PDF
31. Rapid Spread in Japan of Unusual G9P[8] Human Rotavirus Strains Possessing NSP4 Genes of E2 Genotype
- Author
-
Saori, Fukuda, Yuki, Akari, Riona, Hatazawa, Manami, Negoro, Takaaki, Tanaka, Kazutoyo, Asada, Haruna, Nakamura, Katsumi, Sugiura, Masakazu, Umemoto, Haruo, Kuroki, Hiroaki, Ito, Shigeki, Tanaka, Mitsue, Ito, Tomihiko, Ide, Takayuki, Murata, Kiyosu, Taniguchi, Shigeru, Suga, Hajime, Kamiya, Takashi, Nakano, Koki, Taniguchi, and Satoshi, Komoto
- Subjects
Rotavirus ,Microbiology (medical) ,Infectious Diseases ,Genotype ,Japan ,Humans ,Genome, Viral ,General Medicine ,Child ,Phylogeny ,Rotavirus Infections - Abstract
The emergence of unusual G9P[8]-E2 human rotaviruses in the Tokyo metropolitan area, Japan, in 2018 has been reported. During rotavirus strain surveillance in different regions of Japan (Mie, Okayama, and Chiba prefectures), G9P[8]-E2 strains were detected in children with diarrhea from all three prefectures. Here, we characterized the whole genome of seven representative G9P[8]-E2 strains. In the full-genome-based analysis, the seven study strains exhibited a unique genotype configuration with the NSP4 gene of genogroup 2 in a genogroup 1 genomic backbone: G9-P[8]-I1-R1-C1-M1-A1-N1-T1-E2-H1. This genotype constellation was shared by the Tokyo G9P[8]-E2 strains. Phylogenetic analysis showed that all 11 genes, except NSP4, of the seven study strains appeared to have originated from co-circulating Wa-like G9P[8]-E1 strains. In contrast, NSP4 appeared to have originated from the co-circulating DS-1-like G2P[4]-E2 strains. Thus, G9P[8]-E2 strains appear to be derived through reassortment between G9P[8]-E1 and G2P[4]-E2 strains in Japan. Notably, the seven study G9P[8]-E2 strains and Tokyo G9P[8]-E2 strains were revealed to have 11-segment genomes almost indistinguishable from one another in their sequences (99.3-100%), indicating all these G9P[8]-E2 strains had a common origin. To our knowledge, this is the first description of the rapid spread of G9P[8]-E2 strains across a country.
- Published
- 2022
- Full Text
- View/download PDF
32. Epstein-Barr Virus BBLF1 Mediates Secretory Vesicle Transport to Facilitate Mature Virion Release
- Author
-
Md Kamal Uddin, Takahiro Watanabe, Masataka Arata, Yoshitaka Sato, Hiroshi Kimura, and Takayuki Murata
- Subjects
Virology ,Insect Science ,Immunology ,Microbiology - Abstract
Several viruses have been linked to the development of cancer in humans. Epstein-Barr virus (EBV), the first identified human oncovirus, causes a wide range of cancers.
- Published
- 2023
- Full Text
- View/download PDF
33. PD-L1 upregulation by lytic induction of Epstein-Barr Virus
- Author
-
Yusuke Yanagi, Yuya Hara, Seiyo Mabuchi, Takahiro Watanabe, Yoshitaka Sato, Hiroshi Kimura, and Takayuki Murata
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,NF-kappa B ,B7-H1 Antigen ,Cell Line ,Up-Regulation ,Viral Proteins ,Epstein-Barr Virus Nuclear Antigens ,Gene Expression Regulation ,Virology ,Gene Knockdown Techniques ,Host-Pathogen Interactions ,Humans ,Proto-Oncogene Proteins c-akt ,Cells, Cultured ,Signal Transduction - Abstract
Epstein-Barr virus (EBV) is an etiologic agent of infectious mononucleosis and several malignancies. Here, we found that reactivation of EBV resulted in increased programmed cell death-ligand 1 (PD-L1) expression in a cell type-dependent manner. Lytic induction in EBV-positive Akata, AGS, MutuI, and Jijoye cell lines increased PD-L1 levels, but cells such as EBV-negative Akata, MutuIII, and P3HR1 did not have increased PD-L1. EBV in the P3HR1 cell line has a deletion in the EBNA2 gene, while EBV in its parental cell line, Jijoye, has the complete EBNA2 gene. PD-L1 expression by lytic induction was reduced when EBNA2 was knocked down. In addition, pharmacological inhibition indicated involvement of nuclear factor kappa B, mitogen-activated protein kinase, and AKT signaling. These results suggest that EBV likely evades immunity by inducing PD-L1 upon reactivation, through the increased expression of EBNA2 and activation of signaling pathways.
- Published
- 2022
34. Data from Anti-CCR4 Monoclonal Antibody Mogamulizumab for the Treatment of EBV-Associated T- and NK-Cell Lymphoproliferative Diseases
- Author
-
Hiroshi Kimura, Takayuki Murata, Fumi Goshima, Yoshinori Ito, Michio Suzuki, Yoshitaka Sato, Mohammed Siddiquey, Seiko Iwata, Yutaka Hiramatsu, and Tetsuhiro Kanazawa
- Abstract
Purpose: Epstein–Barr virus (EBV) infects not only B cells but also T cells and natural killer (NK) cells, and T- and NK-cell lymphoproliferative diseases (T/NK-LPD) that are refractory to conventional chemotherapies may develop. To identify a molecular-targeted therapy for EBV-associated T/NK-LPDs, we investigated whether CC chemokine receptor 4 (CCR4) was expressed on EBV-infected T and/or NK cells and whether a humanized anti-CCR4 monoclonal antibody, mogamulizumab, was effective.Experimental Design: CCR4 expression was examined in various cell lines. In vitro, the effects of mogamulizumab on cell lines were evaluated in the presence of peripheral blood mononuclear cells from volunteers. In vivo, the effects of mogamulizumab were evaluated using a murine xenograft model. CCR4 expression was examined on EBV-infected cells from patients with EBV-associated T/NK-LPDs. Ex vivo, the effects of mogamulizumab were evaluated using patient lymphocytes.Results: CCR4 expression was confirmed in most EBV-positive T and NK cell lines. Mogamulizumab induced antibody-dependent cellular cytotoxicity (ADCC) activity against CCR4-positive cell lines, and inhibited the growth of EBV-positive NK-cell lymphomas in a murine xenograft model. Furthermore, CCR4 was expressed on EBV-infected cells in 8 of 17 patients with EBV-associated T/NK-LPDs. Interestingly, CCR4 was positive in 5 of 5 patients with hydroa vacciniforme, a photodermatosis caused by the clonal expansion of EBV-infected γδT cells. EBV-positive γδT cells were obtained from a patient with hydroa vacciniforme and subjected to an antibody-dependent cell-mediated cytotoxicity (ADCC) assay. The γδT cells that were positive for CCR4 were killed by mogamulizumab via ADCC.Conclusions: These results indicate that mogamulizumab may be a therapeutic option against EBV-associated T/NK-LPDs. Clin Cancer Res; 20(19); 5075–84. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
35. Supplementary Table 1 from Anti-CCR4 Monoclonal Antibody Mogamulizumab for the Treatment of EBV-Associated T- and NK-Cell Lymphoproliferative Diseases
- Author
-
Hiroshi Kimura, Takayuki Murata, Fumi Goshima, Yoshinori Ito, Michio Suzuki, Yoshitaka Sato, Mohammed Siddiquey, Seiko Iwata, Yutaka Hiramatsu, and Tetsuhiro Kanazawa
- Abstract
Summary of patients with EBV-associated lymphoproliferative diseases and CCR4 expression.
- Published
- 2023
- Full Text
- View/download PDF
36. N6-methyladenosine Modification of Hepatitis B Virus RNA in the Coding Region of HBx
- Author
-
Takayuki Murata, Satoko Iwahori, Yusuke Okuno, Hironori Nishitsuji, Yusuke Yanagi, Koichi Watashi, Takaji Wakita, Hiroshi Kimura, and Kunitada Shimotohno
- Subjects
Inorganic Chemistry ,m6A modification ,primary hepatocyte ,Organic Chemistry ,HBV ,RNA ,General Medicine ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Catalysis ,Computer Science Applications - Abstract
N6-methyladenosine (m6A) is a post-transcriptional modification of RNA involved in transcript transport, degradation, translation, and splicing. We found that HBV RNA is modified by m6A predominantly in the coding region of HBx. The mutagenesis of methylation sites reduced the HBV mRNA and HBs protein levels. The suppression of m6A by an inhibitor or knockdown in primary hepatocytes decreased the viral RNA and HBs protein levels in the medium. These results suggest that the m6A modification of HBV RNA is needed for the efficient replication of HBV in hepatocytes.
- Published
- 2023
- Full Text
- View/download PDF
37. A STING inhibitor suppresses EBV‐induced B cell transformation and lymphomagenesis
- Author
-
Takahiro Watanabe, Keisuke Mantoku, Shouhei Miyagi, H. M. Abdullah Al Masud, Yusuke Yanagi, Shigeo Nakamura, Kamal Uddin, Hiroshi Kimura, Takeshi Suzuki, Masataka Arata, Takayuki Murata, Yasuyuki Miyake, Jun-ichi Kawada, Yoshitaka Sato, Yuya Hara, and Ken Sago
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Cancer Research ,PAMPs ,Lymphoma, B-Cell ,Cell Survival ,T-Lymphocytes ,Antineoplastic Agents ,Epstein‐Barr virus ,medicine.disease_cause ,Inflammation and Virology ,Peripheral blood mononuclear cell ,Jurkat Cells ,Mice ,Interferon ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,B cell ,Cell Proliferation ,Innate immune system ,Chemistry ,Membrane Proteins ,Original Articles ,General Medicine ,Cell Transformation, Viral ,Survival Analysis ,Xenograft Model Antitumor Assays ,Epstein–Barr virus ,EBV‐LPD ,Sting ,HEK293 Cells ,Treatment Outcome ,medicine.anatomical_structure ,Oncology ,Stimulator of interferon genes ,Cancer research ,Original Article ,Signal transduction ,cGAS ,STING ,medicine.drug - Abstract
Epstein‐Barr virus‐associated lymphoproliferative disease (EBV‐LPD) is frequently fatal. Innate immunity plays a key role in protecting against pathogens and cancers. The stimulator of interferon genes (STING) is regarded as a key adaptor protein allowing DNA sensors recognizing exogenous cytosolic DNA to activate the type I interferon signaling cascade. In terms of EBV tumorigenicity, the role of STING remains elusive. Here we showed that treatment with the STING inhibitor, C‐176, suppressed EBV‐induced transformation in peripheral blood mononuclear cells. In an EBV‐LPD mouse model, C‐176 treatment also inhibited tumor formation and prolonged survival. Treatment with B cells alone did not affect EBV transformation, but suppression of EBV‐induced transformation was observed in the presence of T cells. Even without direct B cell‐T cell contact in a transwell system, the inhibitor reduced the transformation activity, indicating that intercellular communication by humoral factors was critical to prevent EBV‐induced transformation. These findings suggest that inhibition of STING signaling pathway with C‐176 could be a new therapeutic target of EBV‐LPD., Treatment with STING inhibitor, C‐176, suppressed the tumor formation in a mouse model of EBV LPDs. Although inhibitor treatment with B cells in vitro alone did not affect EBV transformation, the suppression of EBV‐induced transformation was observed in the presence of T cells
- Published
- 2021
- Full Text
- View/download PDF
38. Virological and genomic analysis of SARS-CoV-2 from a favipiravir clinical trial cohort
- Author
-
Yohei Doi, Chang Kweng Lim, Ayumi Shintani, Masahiro Suzuki, Takumi Imai, Aki Sakurai, Takayuki Murata, Tomihiko Ide, and Satoshi Komoto
- Subjects
Male ,0301 basic medicine ,Microbiology (medical) ,China ,viruses ,030106 microbiology ,Context (language use) ,Favipiravir ,Antiviral Agents ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Pharmacotherapy ,Japan ,Randomized controlled trial ,law ,Humans ,Medicine ,Pharmacology (medical) ,030212 general & internal medicine ,Genome epidemiology ,Cytopathic effect ,SARS-CoV-2 ,business.industry ,COVID-19 ,Genomics ,Amides ,Virology ,Europe ,Clinical trial ,Treatment Outcome ,Infectious Diseases ,Pyrazines ,Cohort ,Original Article ,business ,Viral load - Abstract
Introduction Several clinical studies have reported the efficacy of favipiravir in reducing viral load and shortening the duration of symptoms. However, the viability of SARS-CoV-2 in the context of favipiravir therapy and the potential for resistance development is unclear. Methods We sequenced SARS-CoV-2 in nasopharyngeal specimens collected from patients who participated in a randomized clinical trial of favipiravir at hospitals across Japan between March and May 2020. Paired genomes were sequenced from those who remained RT-PCR-positive 5–8 days into favipiravir therapy. Daily nasopharyngeal specimens from 69 patients who were RT-PCR-positive at randomization were examined for a cytopathic effect (CPE). Results Some strains early in the trial belonged to clade 19 B, whereas the majority belonged to clade 20 B. The median time from the disease onset to negative CPE was 9 days. CPE was strongly correlated with the time from disease onset, viral load, age, and male sex. Among 23 patients for whom paired genomes were available, all except one had identical genomes. Two mutations were observed in one patient who received favipiravir, neither in the RdRp gene. Conclusions The SARS-CoV-2 genome distribution in this clinical trial conducted in Japan reflected the early influx of strains from China followed by replacement by strains from Europe. CPE was significantly associated with age, male sex, and viral loads but not with favipiravir therapy. There was no evidence of resistance development during favipiravir therapy.
- Published
- 2021
- Full Text
- View/download PDF
39. Full genome-based characterization of G4P[6] rotavirus strains from diarrheic patients in Thailand: Evidence for independent porcine-to-human interspecies transmission events
- Author
-
Ratana Tacharoenmuang, Busarawan Sriwanthana, Yoshiki Kawamura, Naokazu Takeda, Ballang Uppapong, Tipsuda Luechakham, Karun Sutthiwarakom, Masashi Tatsumi, Napa Onvimala, Sompong Upachai, Satoshi Komoto, Santip Kongjorn, Tomihiko Ide, Tetsushi Yoshikawa, Riona Hatazawa, Koki Taniguchi, Saori Fukuda, Phakapun Singchai, Takayuki Murata, Kazushi Motomura, Ratigorn Guntapong, and Kriangsak Ruchusatsawast
- Subjects
Diarrhea ,Rotavirus ,medicine.medical_specialty ,Swine ,viruses ,Genome, Viral ,Biology ,medicine.disease_cause ,Genome ,Rotavirus Infections ,03 medical and health sciences ,Medical microbiology ,Species Specificity ,Virology ,Genotype ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Gene ,Phylogeny ,030304 developmental biology ,Swine Diseases ,0303 health sciences ,NSP1 ,Phylogenetic tree ,030306 microbiology ,Transmission (medicine) ,virus diseases ,General Medicine - Abstract
The exact evolutionary patterns of human G4P[6] rotavirus strains remain to be elucidated. Such strains possess unique and strain-specific genotype constellations, raising the question of whether G4P[6] strains are primarily transmitted via independent interspecies transmission or human-to-human transmission after interspecies transmission. Two G4P[6] rotavirus strains were identified in fecal specimens from hospitalized patients with severe diarrhea in Thailand, namely, DU2014-259 (RVA/Human-wt/THA/DU2014-259/2014/G4P[6]) and PK2015-1-0001 (RVA/Human-wt/THA/PK2015-1-0001/2015/G4P[6]). Here, we analyzed the full genomes of the two human G4P[6] strains, which provided the opportunity to study and confirm their evolutionary origin. On whole genome analysis, both strains exhibited a unique Wa-like genotype constellation of G4-P[6]-I1-R1-C1-M1-A8-N1-T1-E1-H1. The NSP1 genotype A8 is commonly found in porcine rotavirus strains. Furthermore, on phylogenetic analysis, each of the 11 genes of strains DU2014-259 and PK2015-1-0001 appeared to be of porcine origin. On the other hand, the two study strains consistently formed distinct clusters for nine of the 11 gene segments (VP4, VP6, VP1-VP3, and NSP2-NSP5), strongly indicating the occurrence of independent porcine-to-human interspecies transmission events. Our observations provide important insights into the origin of zoonotic G4P[6] strains, and into the dynamic interaction between porcine and human rotavirus strains.
- Published
- 2021
- Full Text
- View/download PDF
40. RNAseq analysis identifies involvement of EBNA2 in PD-L1 induction during Epstein-Barr virus infection of primary B cells
- Author
-
H. M. Abdullah Al Masud, Takayuki Murata, Hiroshi Kimura, Takahiro Watanabe, Yusuke Yanagi, Yusuke Okuno, Yohei Narita, Teru Kanda, and Yoshitaka Sato
- Subjects
Herpesvirus 4, Human ,Programmed cell death ,B7-H1 Antigen ,Virus ,Viral Proteins ,03 medical and health sciences ,hemic and lymphatic diseases ,Virology ,PD-L1 ,medicine ,Humans ,Gene ,Epstein–Barr virus infection ,Cells, Cultured ,B cell ,030304 developmental biology ,B-Lymphocytes ,0303 health sciences ,biology ,Sequence Analysis, RNA ,030302 biochemistry & molecular biology ,Promoter ,Cell cycle ,medicine.disease ,Molecular biology ,HEK293 Cells ,medicine.anatomical_structure ,Epstein-Barr Virus Nuclear Antigens ,biology.protein - Abstract
Epstein-Barr virus (EBV) is a causative agent of infectious mononucleosis and several types of malignancy. RNAseq of peripheral blood primary B cell samples infected with wild-type EBV revealed that expression of programmed cell death ligand-1 (PD-L1) is markedly induced by infection. This induction of PD-L1 was alleviated by knockout of the EBNA2 gene, but knockout of LMP1 had little effect. ChIPseq, ChIA-PET, and reporter assays further confirmed that EBNA2-binding sites in the promoter region and at 130 kb downstream of the PD-L1 gene played important roles in PD-L1 induction. Our results indicate that EBV mainly utilizes the EBNA2 gene for induction of PD-L1 and to evade host immunity on infection of primary B cells. Furthermore, pathway analysis revealed that genes involved in the cell cycle, metabolic processes, membrane morphogenesis, and vesicle regulation were induced by EBNA2, and that EBNA2 suppressed genes related to immune signaling.
- Published
- 2021
- Full Text
- View/download PDF
41. Comprehensive Analyses of Intraviral Epstein-Barr Virus Protein–Protein Interactions Hint Central Role of BLRF2 in the Tegument Network
- Author
-
Yuya Hara, Takahiro Watanabe, Masahiro Yoshida, H. M. Abdullah Al Masud, Hiromichi Kato, Tomohiro Kondo, Reiji Suzuki, Shutaro Kurose, Md. Kamal Uddin, Masataka Arata, Shouhei Miyagi, Yusuke Yanagi, Yoshitaka Sato, Hiroshi Kimura, and Takayuki Murata
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Viral Proteins ,Structure and Assembly ,Virology ,Insect Science ,Immunology ,Humans ,Protein Interaction Maps ,Virus Replication ,Microbiology - Abstract
Protein–protein interactions (PPIs) are crucial for various biological processes. Epstein-Barr virus (EBV) proteins typically form complexes, regulating the replication and persistence of the viral genome in human cells. However, the role of EBV protein complexes under physiological conditions remains unclear. In this study, we performed comprehensive analyses of EBV PPIs in living cells using the NanoBiT system. We identified 195 PPIs, many of which have not previously been reported. Computational analyses of these PPIs revealed that BLRF2, which is only found in gammaherpesviruses, is a central protein in the structural network of EBV tegument proteins. To characterize the role of BLRF2, we generated two BLRF2 knockout EBV clones using CRISPR/Cas9. BLRF2 knockout significantly decreased the production of infectious virus particles, which was partially restored by exogenous BLRF2 expression. In addition, self-association of BLRF2 protein was found, and mutation of the residues crucial for the self-association affected stability of the protein. Our data imply that BLRF2 is a tegument network hub that plays important roles in progeny virion maturation. IMPORTANCE EBV remains a significant public health challenge, causing infectious mononucleosis and several cancer types. Therefore, the better understanding of the molecular mechanisms underlying EBV replication is of high clinical importance. As protein–protein interactions (PPIs) are major regulators of virus-associated pathogenesis, comprehensive analyses of PPIs are essential. Previous studies on PPIs in EBV or other herpesviruses have predominantly employed the yeast two-hybrid (Y2H) system, immunoprecipitation, and pulldown assays. Herein, using a novel luminescence-based method, we identified 195 PPIs, most of which have not previously been reported. Computational and functional analyses using knockout viruses revealed that BLRF2 plays a central role in the EBV life cycle, which makes it a valuable target for drug development.
- Published
- 2022
- Full Text
- View/download PDF
42. Rotavirus incapable of NSP6 expression can cause diarrhea in suckling mice
- Author
-
Saori Fukuda, Masanori Kugita, Yuki Higashimoto, Kazuya Shiogama, Hanako Tsujikawa, Kyoko Moriguchi, Naoto Ito, Makoto Sugiyama, Shizuko Nagao, Takayuki Murata, Koki Taniguchi, and Satoshi Komoto
- Subjects
Diarrhea ,Rotavirus ,Mice ,Virology ,Animals ,Cells, Cultured ,Rotavirus Infections ,Cell Line - Abstract
The group A rotavirus (RVA) genome comprising 11 double-stranded RNAs encodes six structural proteins (VP1-VP4, VP6, and VP7) and six non-structural proteins (NSP1-NSP6). Among these 12 rotaviral proteins, NSP6 has been less studied as to its function. We previously prepared a recombinant NSP6-deficient RVA derived from simian strain SA11-L2 by reverse genetics, and found that the NSP6-deficient virus grew well in cell culture, although its growth was less abundant than that of the parental SA11-L2 strain. In this study, we examined the potency of a recombinant RVA incapable of NSP6 expression to cause diarrhoea in suckling mice. The suckling mice infected with the NSP6-deficient virus apparently experienced diarrhoea, although the symptom was milder and the duration of diarrhoea was shorter than in the mice infected with the authentic SA11-L2 strain. Thus, together with the results obtained for cultured cells in the previous study, it can be concluded that NSP6 is not necessarily required for replication and pathogenicity in vitro and in vivo.
- Published
- 2022
43. Serine 13 of the human cytomegalovirus viral cyclin-dependent kinase UL97 is required for regulatory protein 14-3-3 binding and UL97 stability
- Author
-
Satoko Iwahori, Angie C. Umaña, Robert F. Kalejta, and Takayuki Murata
- Subjects
Proteomics ,Phosphotransferases (Alcohol Group Acceptor) ,14-3-3 Proteins ,Serine ,Humans ,Cytomegalovirus ,Cell Biology ,Molecular Biology ,Biochemistry ,Cyclin-Dependent Kinases - Abstract
The human cytomegalovirus (HCMV) UL97 protein is a conserved herpesvirus protein kinase (CHPK) and a viral cyclin-dependent kinase (v-CDK). However, mechanisms regulating its activity in the context of infection are unknown. Here, we identified several cellular regulatory 14-3-3 proteins as UL97-interacting partners that promote UL97 stability. Humans are known to encode seven isoforms of 14-3-3 proteins (β, ε, η, γ, σ, θ, and ζ) that bind phosphoserines or phosphothreonines to impact protein structure, stability, activity, and localization. Our proteomic analysis of UL97 identified 49 interacting partners, including 14-3-3 isoforms β, η, and γ. Furthermore, coimmunoprecipitation with Western blotting assays demonstrated that UL97 interaction with 14-3-3 isoforms β, ε, η, γ, and θ occurs in a kinase activity-dependent manner. Using mutational analysis, we determined the serine residue at amino acid 13 of UL97 is crucial for 14-3-3 interaction. We demonstrate UL97 S13A (serine to alanine substitution at residue 13) retains kinase activity but the mutant protein accumulated at lower levels than WT UL97. Finally, we show both laboratory (AD169) and clinical (TB40/E) strains of HCMV encoding UL97 S13A replicated with WT kinetics in fibroblasts but showed decreased UL97 accumulation. Taken together, we conclude that 14-3-3 proteins interact with and stabilize UL97 during HCMV infection.
- Published
- 2022
44. Estrogen induces the expression of EBV lytic protein ZEBRA, a marker of poor prognosis in nasopharyngeal carcinoma
- Author
-
Hirotomo Dochi, Satoru Kondo, Takayuki Murata, Masaki Fukuyo, Asuka Nanbo, Kousho Wakae, Wen‐Ping Jiang, Toshihide Hamabe‐Horiike, Mariko Tanaka, Takumi Nishiuchi, Harue Mizokami, Makiko Moriyama‐Kita, Eiji Kobayashi, Nobuyuki Hirai, Takeshi Komori, Takayoshi Ueno, Yosuke Nakanishi, Miyako Hatano, Kazuhira Endo, Hisashi Sugimoto, Naohiro Wakisaka, Shin‐Hun Juang, Masamichi Muramatsu, Atsushi Kaneda, and Tomokazu Yoshizaki
- Subjects
Cancer Research ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Aromatase ,Nasopharyngeal Carcinoma ,Oncology ,Estrogen Receptor alpha ,Trans-Activators ,Humans ,Estrogens ,Nasopharyngeal Neoplasms ,General Medicine - Abstract
Several epidemiological studies have suggested that Epstein-Barr virus (EBV) lytic infection is essential for the development of nasopharyngeal carcinoma (NPC), as the elevation of antibody titers against EBV lytic proteins is a common feature of NPC. Although ZEBRA protein is a key trigger for the initiation of lytic infection, whether its expression affects the prognosis and pathogenesis of NPC remains unclear. In this study, 64 NPC biopsy specimens were analyzed using immunohistochemistry. We found that ZEBRA was significantly associated with a worsening of progression-free survival in NPC (adjusted hazard ratio, 3.58; 95% confidence interval, 1.08-11.87; p = 0.037). Moreover, ZEBRA expression positively correlated with key endocrinological proteins, estrogen receptor α, and aromatase. The transcriptional level of ZEBRA is activated by estrogen in an estrogen receptor α-dependent manner, resulting in an increase in structural gene expression levels and extracellular virus DNA copy number in NPC cell lines, reminiscent of lytic infection. Interestingly, it did not suppress cellular proliferation or increase apoptosis, in contrast with cells treated with 12-O-tetradecanoylphorbol-13-acetate and sodium butyrate, indicating that viral production induced by estrogen is not a cell lytic phenomenon. Our results suggest that intratumoral estrogen overproduced by aromatase could induce ZEBRA expression and EBV reactivation, contributing to the progression of NPC.
- Published
- 2022
45. Development of the electric wheelchair hands-free semi-automatic control system using the surface-electromyogram of facial muscles.
- Author
-
Hiroki Tamura, Takayuki Murata, Yuki Yamashita, Koichi Tanno, and Yasufumi Fuse
- Published
- 2012
- Full Text
- View/download PDF
46. Rapid generation of rotavirus single-gene reassortants by means of eleven plasmid-only based reverse genetics
- Author
-
Riona Hatazawa, Koki Taniguchi, Takayuki Murata, Tetsushi Yoshikawa, Saori Fukuda, Yoshiki Kawamura, and Satoshi Komoto
- Subjects
Rotavirus ,0301 basic medicine ,DNA, Complementary ,Genotype ,Swine ,030106 microbiology ,Reassortment ,Genome, Viral ,Biology ,medicine.disease_cause ,Genome ,Rotavirus Infections ,Virus ,Cell Line ,03 medical and health sciences ,Plasmid ,Cricetinae ,Virology ,medicine ,Animals ,Humans ,Gene ,Recombination, Genetic ,Genetics ,Haplorhini ,Reverse Genetics ,Reverse genetics ,030104 developmental biology ,RNA, Viral ,Capsid Proteins ,Reassortant Viruses ,Plasmids - Abstract
Reassortment is an important mechanism in the evolution of group A rotaviruses (RVAs), yielding viruses with novel genetic and phenotypic traits. The classical methods for generating RVA reassortants with the desired genetic combinations are laborious and time-consuming because of the screening and selection processes required to isolate a desired reassortant. Taking advantage of a recently developed RVA reverse genetics system based on just 11 cloned cDNAs encoding the RVA genome (11 plasmid-only system), we prepared a panel of simian SA11-L2 virus-based single-gene reassortants, each containing 1 segment derived from human KU virus of the G1P[8] genotype. It was shown that there was no gene-specific restriction of the reassortment potential. In addition to these 11 single-gene reassortants, a triple-gene reassortant with KU-derived core-encoding VP1–3 gene segments with the SA11-L2 genetic background, which make up a virion composed of the KU-based core, and SA11-L2-based intermediate and outer layers, could also be prepared with the 11 plasmid-only system. Finally, for possible clinical application of this system, we generated a series of VP7 reassortants representing all the major human RVA G genotypes (G1–4, G9 and G12) efficiently. The preparation of each of these single-gene reassortants was achieved within just 2 weeks. Our results demonstrate that the 11 plasmid-only system allows the rapid and reliable generation of RVA single-gene reassortants, which will be useful for basic research and clinical applications.
- Published
- 2020
- Full Text
- View/download PDF
47. Reverse genetics system for human rotaviruses
- Author
-
Koki Taniguchi, Saori Fukuda, Takayuki Murata, and Satoshi Komoto
- Subjects
Rotavirus ,0303 health sciences ,DNA, Complementary ,030306 microbiology ,viruses ,Immunology ,Genome, Viral ,Biology ,Microbiology ,Virology ,Reverse Genetics ,Reverse genetics ,Virus ,Viral vector ,Viral Proteins ,03 medical and health sciences ,Complementary DNA ,Human rotavirus ,Animals ,Humans ,RNA, Viral ,Plasmids ,030304 developmental biology - Abstract
A reverse genetics technology is an incredibly useful technique both for a proper understanding of different aspects of virus biology and for the generation of complementary DNA (cDNA)-derived infectious viruses, which can act as safe and effective vaccines and viral vectors. Rotaviruses (RVAs), especially human RVAs (HuRVAs), had been very refractory to this technology until very recently. Here, we describe the historical background of the development of a long-awaited HuRVA reverse genetics system, culminating in the generation of replicative HuRVAs entirely from cloned cDNAs.
- Published
- 2020
- Full Text
- View/download PDF
48. EBV genome variations enhance clinicopathological features of nasopharyngeal carcinoma in a non-endemic region
- Author
-
Satoru Kondo, Yusuke Okuno, Takayuki Murata, Hirotomo Dochi, Naohiro Wakisaka, Harue Mizokami, Makiko Moriyama‐Kita, Eiji Kobayashi, Makoto Kano, Takeshi Komori, Nobuyuki Hirai, Takayoshi Ueno, Yosuke Nakanishi, Kazuhira Endo, Hisashi Sugimoto, Hiroshi Kimura, and Tomokazu Yoshizaki
- Subjects
Cancer Research ,China ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Nasopharyngeal Carcinoma ,Oncology ,Humans ,Nasopharyngeal Neoplasms ,General Medicine ,Genome, Viral - Abstract
Nasopharyngeal carcinoma (NPC) is caused by infection with Epstein-Barr virus (EBV) and endemic in certain geographic regions. EBV lytic gene, BALF2, closely associates with viral reactivation and BALF2 gene variation, the H-H-H strain, causes NPC in endemic region, southern China. Here, we investigate whether such EBV variations also affect NPC in a non-endemic region, Japan. Viral genome sequencing with 47 EBV isolates of Japanese NPC were performed and compared with those of other EBV-associated diseases from Japan or NPC in Southern China. EBV genomes of Japanese NPC are different from those of other diseases in Japan or endemic NPC; Japanese NPC was not affected by the endemic strain (the BALF2 H-H-H) but frequently carried the type 2 EBV or the strain with intermediate risk of endemic NPC (the BALF2 H-H-L). Seven single nucleotide variations were specifically associated with Japanese NPC, of which six were present in both type 1 and 2 EBV genomes, suggesting the contribution of the type 2 EBV-derived haplotype. This observation was supported by a higher viral titer and stronger viral reactivation in NPC with either type 2 or H-H-L strains. Our results highlight the importance of viral strains and viral reactivation in the pathogenesis of non-endemic NPC.
- Published
- 2022
49. Molecular Basis of Epstein-Barr Virus Latency Establishment and Lytic Reactivation
- Author
-
Tomoki Inagaki, Hiroshi Kimura, Yoshitaka Sato, Takayuki Murata, Atsuko Sugimoto, Yusuke Yanagi, and Takahiro Watanabe
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,reactivation ,viruses ,Review ,Biology ,medicine.disease_cause ,Microbiology ,Virus ,Immediate-Early Proteins ,EBV ,oncogenesis ,Virology ,Gene expression ,medicine ,Humans ,Gene ,latency ,epigenetics ,Germinal center ,Epstein–Barr virus ,QR1-502 ,BZLF1 ,Virus Latency ,Infectious Diseases ,Lytic cycle ,Trans-Activators ,Virus Activation ,Carcinogenesis ,transcription - Abstract
Epstein–Barr virus (EBV) is a causative agent of infectious mononucleosis and several types of cancer. Like other herpesviruses, it establishes an asymptomatic, life-long latent infection, with occasional reactivation and shedding of progeny viruses. During latency, EBV expresses a small number of viral genes, and exists as an episome in the host–cell nucleus. Expression patterns of latency genes are dependent on the cell type, time after infection, and milieu of the cell (e.g., germinal center or peripheral blood). Upon lytic induction, expression of the viral immediate-early genes, BZLF1 and BRLF1, are induced, followed by early gene expression, viral DNA replication, late gene expression, and maturation and egress of progeny virions. Furthermore, EBV reactivation involves more than just progeny production. The EBV life cycle is regulated by signal transduction, transcription factors, promoter sequences, epigenetics, and the 3D structure of the genome. In this article, the molecular basis of EBV latency establishment and reactivation is summarized.
- Published
- 2021
50. Antitumor activity of cyclin‐dependent kinase inhibitor alsterpaullone in Epstein‐Barr virus‐associated lymphoproliferative disorders
- Author
-
Fumi Goshima, Yoshitaka Sato, Hiroki Matsuda, Yusuke Yanagi, Takahiro Watanabe, Takayuki Murata, Masahiro Yoshida, H. M. Abdullah Al Masud, Yuya Hara, Masahiro Takayama, and Hiroshi Kimura
- Subjects
0301 basic medicine ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Cancer Research ,Indoles ,CDK ,Lymphoproliferative disorders ,Antineoplastic Agents ,Apoptosis ,medicine.disease_cause ,Inflammation and Virology ,Virus ,Cell Line ,Mice ,03 medical and health sciences ,rituximab ,0302 clinical medicine ,EBV ,Mice, Inbred NOD ,Cyclin-dependent kinase ,hemic and lymphatic diseases ,Tumor Virus ,medicine ,Animals ,Humans ,Protein Kinase Inhibitors ,biology ,Kinase ,Chemistry ,Cyclin-Dependent Kinase 2 ,Cyclin-dependent kinase 2 ,G1 Phase ,Original Articles ,Cell Cycle Checkpoints ,General Medicine ,Benzazepines ,medicine.disease ,Epstein–Barr virus ,Lymphoproliferative Disorders ,HEK293 Cells ,030104 developmental biology ,LPD ,Oncology ,vPIC ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Original Article - Abstract
Epstein‐Barr virus (EBV) is a well‐established tumor virus that has been implicated in a wide range of immunodeficiency‐associated lymphoproliferative disorders (LPDs). Although rituximab, a CD20 mAb, has proven effective against EBV‐associated LPDs, prolonged use of this drug could lead to resistance due to the selective expansion of CD20− cells. We have previously shown that cyclin‐dependent kinase (CDK) inhibitors are able to specifically suppress the expression of viral late genes, particularly those encoding structural proteins; however, the therapeutic effect of CDK inhibitors against EBV‐associated LPDs is not clear. In this study, we examined whether CDK inhibitors confer a therapeutic effect against LPDs in vivo. Treatment with alsterpaullone, an inhibitor of the CDK2 complex, resulted in a survival benefit and suppressed tumor invasion in a mouse model of LPDs. Inhibition of CDK efficiently induced G1 cell cycle arrest and apoptosis in EBV‐positive B cells. These results suggest that alsterpaullone suppresses cell cycle progression, resulting in the antitumor effect observed in vivo., Epstein‐Barr virus (EBV) is a well‐established tumor virus that has been implicated in a wide range of immunodeficiency‐associated lymphoproliferative disorders (LPDs). In this study, we examined whether cyclin‐dependent kinase (CDK) inhibitors confer a therapeutic effect against LPDs in vivo. Treatment with alsterpaullone, an inhibitor of the CDK2 complex, resulted in a survival benefit and suppressed tumor invasion in a mouse model of LPDs. Inhibition of CDK efficiently induced G1 cell cycle arrest and apoptosis in EBV‐positive B cells.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.