69 results on '"Seiya Yamayoshi"'
Search Results
2. Induction of IGHV3-53 public antibodies with broadly neutralising activity against SARS-CoV-2 including Omicron subvariants in a Delta breakthrough infection caseResearch in context
- Author
-
Takeo Kuwata, Yu Kaku, Shashwata Biswas, Kaho Matsumoto, Mikiko Shimizu, Yoko Kawanami, Ryuta Uraki, Kyo Okazaki, Rumi Minami, Yoji Nagasaki, Mami Nagashima, Isao Yoshida, Kenji Sadamasu, Kazuhisa Yoshimura, Mutsumi Ito, Maki Kiso, Seiya Yamayoshi, Masaki Imai, Terumasa Ikeda, Kei Sato, Mako Toyoda, Takamasa Ueno, Takako Inoue, Yasuhito Tanaka, Kanako Tarakado Kimura, Takao Hashiguchi, Yukihiko Sugita, Takeshi Noda, Hiroshi Morioka, Yoshihiro Kawaoka, Shuzo Matsushita, Jumpei Ito, Naoko Misawa, Arnon Plianchaisuk, Ziyi Guo, Alfredo Hina, Jr., Keiya Uriu, Kaoru Usui, Wilaiporn Saikruang, Spyridon Lytras, Ryo Yoshimura, Shusuke Kawakubo, Luca Nishimura, Yusuke Kosugi, Shigeru Fujita, Luo Chen, Jarel Elgin M. Tolentino, Lin Pan, Wenye Li, Maximilian Stanley Yo, Kio Horinaka, Mai Suganami, Adam P. Strange, Mika Chiba, Keiko Iida, Naomi Ohsumi, Kaho Okumura, Shiho Tanaka, Eiko Ogawa, Kyoko Yasuda, Tsuki Fukuda, Rina Osujo, Takasuke Fukuhara, Tomokazu Tamura, Rigel Suzuki, Saori Suzuki, Hayato Ito, Keita Matsuno, Hirofumi Sawa, Naganori Nao, Shinya Tanaka, Masumi Tsuda, Lei Wang, Yoshikata Oda, Zannatul Ferdous, Kenji Shishido, Keita Mizuma, Isshu Kojima, Jingshu Li, Tomoya Tsubo, Shuhei Tsujino, So Nakagawa, Kotaro Shirakawa, Akifumi Takaori-Kondo, Kayoko Nagata, Ryosuke Nomura, Yoshihito Horisawa, Yusuke Tashiro, Yugo Kawai, Kazuo Takayama, Rina Hashimoto, Sayaka Deguchi, Yukio Watanabe, Ayaka Sakamoto, Naoko Yasuhara, Tateki Suzuki, Kanako Kimura, Jiei Sasaki, Yukari Nakajima, Hisano Yajima, Yoshitaka Nakata, Hiroki Futatsusako, Takashi Irie, Ryoko Kawabata, Kaori Tabata, Hesham Nasser, Ryo Shimizu, MST Monira Begum, Michael Jonathan, Yuka Mugita, Otowa Takahashi, Kimiko Ichihara, Chihiro Motozono, Sharee Leong, Akatsuki Saito, Maya Shofa, Yuki Shibatani, Tomoko Nishiuchi, Hiroyuki Asakura, Jiri Zahradnik, Prokopios Andrikopoulos, Miguel Padilla-Blanco, and Aditi Konar
- Subjects
SARS-CoV-2 ,Neutralising antibody ,Variant ,Public antibody ,Medicine ,Medicine (General) ,R5-920 - Abstract
Summary: Background: Emergence of SARS-CoV-2 variants that escape neutralising antibodies hampers the development of vaccines and therapeutic antibodies against SARS-CoV-2. IGHV3-53/3-66-derived public antibodies, which are generally specific to the prototype virus and are frequently induced in infected or vaccinated individuals, show minimal affinity maturation and high potency against prototype SARS-CoV-2. Methods: Monoclonal antibodies isolated from a Delta breakthrough infection case were analysed for cross-neutralising activities against SARS-CoV-2 variants. The broadly neutralising antibody K4-66 was further analysed in a hamster model, and the effect of somatic hypermutations was assessed using the inferred germline precursor. Findings: Antibodies derived from IGHV3-53/3-66 showed broader neutralising activity than antibodies derived from IGHV1-69 and other IGHV genes. IGHV3-53/3-66 antibodies neutralised the Delta variant better than the IGHV1-69 antibodies, suggesting that the IGHV3-53/3-66 antibodies were further maturated by Delta breakthrough infection. One IGHV3-53/3-66 antibody, K4-66, neutralised all Omicron subvariants tested, including EG.5.1, BA.2.86, and JN.1, and decreased the viral load in the lungs of hamsters infected with Omicron subvariant XBB.1.5. The importance of somatic hypermutations was demonstrated by the loss of neutralising activity of the inferred germline precursor of K4-66 against Beta and Omicron variants. Interpretation: Broadly neutralising IGHV3-53/3-66 antibodies have potential as a target for the development of effective vaccines and therapeutic antibodies against newly emerging SARS-CoV-2 variants. Funding: This work was supported by grants from AMED (JP23ym0126048, JP22ym0126048, JP21ym0126048, JP23wm0125002, JP233fa627001, JP223fa627009, JP24jf0126002, and JP22fk0108572), and the JSPS (JP21H02970, JK23K20041, and JPJSCCA20240006).
- Published
- 2024
- Full Text
- View/download PDF
3. Age-Stratified Seroprevalence of SARS-CoV-2 Antibodies before and during the Vaccination Era, Japan, February 2020–March 2022
- Author
-
Seiya Yamayoshi, Kiyoko Iwatsuki-Horimoto, Moe Okuda, Michiko Ujie, Atsuhiro Yasuhara, Jurika Murakami, Calvin Duong, Taiki Hamabata, Mutsumi Ito, Shiho Chiba, Ryo Kobayashi, Satoshi Takahashi, Keiko Mitamura, Masao Hagihara, Akimichi Shibata, Yoshifumi Uwamino, Naoki Hasegawa, Toshiaki Ebina, Akihiko Izumi, Hideaki Kato, Hideaki Nakajima, Norio Sugaya, Yuki Seki, Asef Iqbal, Isamu Kamimaki, Masahiko Yamazaki, Yoshihiro Kawaoka, and Yuki Furuse
- Subjects
COVID-19 ,coronavirus disease ,SARS-CoV-2 ,severe acute respiratory syndrome coronavirus 2 ,viruses ,respiratory infections ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
Japan has reported a relatively small number of COVID-19 cases. Because not all infected persons receive diagnostic tests for COVID-19, the reported number must be lower than the actual number of infections. We assessed SARS-CoV-2 seroprevalence by analyzing >60,000 samples collected in Japan (Tokyo Metropolitan Area and Hokkaido Prefecture) during February 2020–March 2022. The results showed that ≈3.8% of the population had become seropositive by January 2021. The seroprevalence increased with the administration of vaccinations; however, among the elderly, seroprevalence was not as high as the vaccination rate. Among children, who were not eligible for vaccination, infection was spread during the epidemic waves caused by the SARS-CoV-2 Delta and Omicron variants. Nevertheless, seroprevalence for unvaccinated children
- Published
- 2022
- Full Text
- View/download PDF
4. Transmission and re-infection of Omicron variant XBB.1.5 in hamstersResearch in context
- Author
-
Peter J. Halfmann, Ryuta Uraki, Makoto Kuroda, Kiyoko Iwatsuki-Horimoto, Seiya Yamayoshi, Mutsumi Ito, and Yoshihiro Kawaoka
- Subjects
XBB.1.5 ,Airborne transmission ,Animal model ,Hamster ,Re-infection ,Medicine ,Medicine (General) ,R5-920 - Abstract
Summary: Background: Like its predecessors in the XBB family, XBB.1.5 is highly immune evasive from therapeutic monoclonal antibodies and neutralizing antibodies generated by vaccination and/or infection. However, there is a lack of in vivo data on XBB.1.5 in animal models such as Syrian hamsters. Methods: Syrian hamsters (females) were used to examine airborne transmission along with virus replication of XBB.1.5 in naïve animals and human ACE2 hamsters with pre-existing immunity from a previous infection with Omicron BA.1. Assays were performed to determine neutralizing antibody responses, and virus titers were determined by standard plaque assays. Findings: Unlike earlier Omicron subvariants, such as BA.1 and BA.2, XBB.1.5 transmitted more efficiently in the hamster model. In addition, XBB.1.5 partially escaped BA.1-immunity from a previous infection with XBB.1.5 replicating in the nasal turbinate tissues and to a lesser extend in the lung tissues of previously infected hamsters. Interpretation: Our in vivo data showing better airborne transmissibility of the Omicron subvariant XBB.1.5 than its predecessor, BA.2, in Syrian hamsters will allow researchers to further investigate amino acid substitutions that give XBB.1.5 a fitness advantage over BA.2 in transmission, data that may be important in studies of SARS-CoV-2 transmission in humans. Funding: This research is supported by grants from the Center for Research on Influenza Pathogenesis and Transmission (CRIPT; 75N93021C00014), funded by the National Institute of Allergy and Infectious Diseases and by a Research Program on Emerging and Reemerging Infectious Diseases (JP21fk0108552 and JP21fk0108615), a Project Promoting Support for Drug Discovery (JP21nf0101632), the Japan Program for Infectious Diseases Research and Infrastructure (JP22wm0125002), and The University of Tokyo Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA) grant (JP223fa627001) from the Japan Agency for Medical Research and Development.
- Published
- 2023
- Full Text
- View/download PDF
5. In SARS-CoV-2 delta variants, Spike-P681R and D950N promote membrane fusion, Spike-P681R enhances spike cleavage, but neither substitution affects pathogenicity in hamstersResearch in context
- Author
-
Yuri Furusawa, Maki Kiso, Shun Iida, Ryuta Uraki, Yuichiro Hirata, Masaki Imai, Tadaki Suzuki, Seiya Yamayoshi, and Yoshihiro Kawaoka
- Subjects
SARS-CoV-2 ,COVID-19 ,Reverse genetics ,Hamster ,Medicine ,Medicine (General) ,R5-920 - Abstract
Summary: Background: The SARS-CoV-2 delta (B.1.617.2 lineage) variant was first identified at the end of 2020 and possessed two unique amino acid substitutions in its spike protein: S-P681R, at the S1/S2 cleavage site, and S-D950N, in the HR1 of the S2 subunit. However, the roles of these substitutions in virus phenotypes have not been fully characterized. Methods: We used reverse genetics to generate Wuhan-D614G viruses with these substitutions and delta viruses lacking these substitutions and explored how these changes affected their viral characteristics in vitro and in vivo. Findings: S-P681R enhanced spike cleavage and membrane fusion, whereas S-D950N slightly promoted membrane fusion. Although S-681R reduced the virus replicative ability especially in VeroE6 cells, neither substitution affected virus replication in Calu-3 cells and hamsters. The pathogenicity of all recombinant viruses tested in hamsters was slightly but not significantly affected. Interpretation: Our observations suggest that the S-P681R and S-D950N substitutions alone do not increase virus pathogenicity, despite of their enhancement of spike cleavage or fusogenicity. Funding: A full list of funding bodies that contributed to this study can be found under Acknowledgments.
- Published
- 2023
- Full Text
- View/download PDF
6. Enhanced Replication of Highly Pathogenic Influenza A(H7N9) Virus in Humans
- Author
-
Seiya Yamayoshi, Maki Kiso, Atsuhiro Yasuhara, Mutsumi Ito, Yuelong Shu, and Yoshihiro Kawaoka
- Subjects
Highly pathogenic ,H7N9 ,PB2 ,PA ,enhanced polymerase activity ,influenza ,Medicine ,Infectious and parasitic diseases ,RC109-216 - Abstract
To clarify the threat posed by emergence of highly pathogenic influenza A(H7N9) virus infection among humans, we characterized the viral polymerase complex. Polymerase basic 2–482R, polymerase basic 2–588V, and polymerase acidic–497R individually or additively enhanced virus polymerase activity, indicating that multiple replication-enhancing mutations in 1 isolate may contribute to virulence.
- Published
- 2018
- Full Text
- View/download PDF
7. A Broadly Reactive Human Anti-hemagglutinin Stem Monoclonal Antibody That Inhibits Influenza A Virus Particle Release
- Author
-
Seiya Yamayoshi, Ryuta Uraki, Mutsumi Ito, Maki Kiso, Sumiho Nakatsu, Atsuhiro Yasuhara, Kohei Oishi, Tadahiro Sasaki, Kazuyoshi Ikuta, and Yoshihiro Kawaoka
- Subjects
Influenza A virus ,Human monoclonal antibody ,HA stem ,Broadly reactive ,Medicine ,Medicine (General) ,R5-920 - Abstract
Many broadly reactive human monoclonal antibodies against the hemagglutinin (HA) stem of influenza A virus have been developed for therapeutic applications. These antibodies typically inhibit viral entry steps, especially the HA conformational change that is required for membrane fusion. To better understand the mechanisms by which such antibodies inhibit viral replication, we established broadly reactive human anti-HA stem antibodies and determined the properties of these antibodies by examining their reactivity with 18 subtypes of HA, evaluating their in vivo protective efficacy, identifying their epitopes, and characterizing their inhibitory mechanisms. Among the eight human monoclonal antibodies we generated, which recognized at least 3 subtypes of the soluble HA antigens tested, clone S9-1-10/5-1 reacted with 18 subtypes of HA and protected mice from lethal infection with H1N1pdm09, H3N2, H5N1, and H7N9 viruses. This antibody recognized the HA2 helix A in the HA stem, and inhibited virus particle release from infected cells but did not block viral entry completely. These results show that broadly reactive human anti-HA stem antibodies can exhibit protective efficacy by inhibiting virus particle release. These findings expand our knowledge of the mechanisms by which broadly reactive stem-targeting antibodies inhibit viral replication and provide valuable information for universal vaccine development.
- Published
- 2017
- Full Text
- View/download PDF
8. Identification of Novel Adjuvants for Ebola Virus-Like Particle Vaccine
- Author
-
Huapeng Feng, Sumiho Nakatsu, Tiago Jose da Silva Lopes, Masaki Imai, Seiya Yamayoshi, Makoto Yamashita, Tokiko Watanabe, and Yoshihiro Kawaoka
- Subjects
Ebola vaccine ,virus-like particle ,adjuvants ,Medicine - Abstract
Ebola virus disease is a severe disease, often fatal, with a mortality rate of up to 90%. Presently, effective treatment and safe prevention options for Ebola virus disease are not available. Therefore, there is an urgent need to develop control measures to prevent or limit future Ebola virus outbreaks. Ebola virus protein-based virus-like particle (VLP) and inactivated whole virion vaccines have demonstrated efficacy in animal models, and the addition of appropriate adjuvants may provide additional benefits to these vaccines, including enhanced immune responses. In this study, we screened 24 compounds from injectable excipients approved for human use in Japan and identified six compounds that significantly enhanced the humoral response to Ebola VLP vaccine in a murine model. Our novel adjuvant candidates for Ebola VLP vaccine have already been demonstrated to be safe when administered intramuscularly or subcutaneously, and therefore, they are closer to clinical trials than adjuvants whose safety profiles are unknown.
- Published
- 2020
- Full Text
- View/download PDF
9. Pathogenesis of Influenza A(H7N9) Virus in Aged Nonhuman Primates
- Author
-
Hiroaki Katsura, Yuriko Tomita, Maki Kiso, Noriko Nakajima, Yoshihiro Kawaoka, Tokiko Watanabe, Jason E. Shoemaker, Hideki Hasegawa, Seiya Yamayoshi, Satoshi Fukuyama, Tadashi Maemura, Robert W. Gregg, Kiyoko Iwatsuki-Horimoto, and Tiago J. S. Lopes
- Subjects
0301 basic medicine ,Aging ,Chemokine ,medicine.medical_treatment ,030106 microbiology ,Inflammation ,Influenza A Virus, H7N9 Subtype ,Virus Replication ,medicine.disease_cause ,Virus ,Proinflammatory cytokine ,Pathogenesis ,Major Articles and Brief Reports ,03 medical and health sciences ,Immune system ,Orthomyxoviridae Infections ,medicine ,Animals ,Immunology and Allergy ,Lung ,biology ,business.industry ,Influenza A virus subtype H5N1 ,Disease Models, Animal ,Macaca fascicularis ,030104 developmental biology ,Infectious Diseases ,Cytokine ,Immunology ,biology.protein ,Cytokines ,Female ,medicine.symptom ,business - Abstract
The avian influenza A(H7N9) virus has caused high mortality rates in humans, especially in the elderly; however, little is known about the mechanistic basis for this. In the current study, we used nonhuman primates to evaluate the effect of aging on the pathogenicity of A(H7N9) virus. We observed that A(H7N9) virus infection of aged animals (defined as age 20–26 years) caused more severe symptoms than infection of young animals (defined as age 2–3 years). In aged animals, lung inflammation was weak and virus infection was sustained. Although cytokine and chemokine expression in the lungs of most aged animals was lower than that in the lungs of young animals, 1 aged animal showed severe symptoms and dysregulated proinflammatory cytokine and chemokine production. These results suggest that attenuated or dysregulated immune responses in aged animals are responsible for the severe symptoms observed among elderly patients infected with A(H7N9) virus.
- Published
- 2020
10. Baloxavir Marboxil Treatment of Nude Mice Infected With Influenza A Virus
- Author
-
Jurika Murakami, Yoshihiro Kawaoka, Maki Kiso, and Seiya Yamayoshi
- Subjects
Dibenzothiepins ,Male ,Pyridones ,Morpholines ,Orthomyxoviridae ,Mutant ,Mice, Nude ,Drug resistance ,medicine.disease_cause ,Antiviral Agents ,Virus ,Mice ,Major Articles and Brief Reports ,Orthomyxoviridae Infections ,Influenza A virus ,medicine ,Animals ,Immunology and Allergy ,Respiratory system ,Dose-Response Relationship, Drug ,biology ,Triazines ,business.industry ,biology.organism_classification ,Virology ,Infectious Diseases ,medicine.anatomical_structure ,biology.protein ,business ,Neuraminidase ,Respiratory tract - Abstract
Background Immunocompromised patients infected with influenza virus require prolonged treatment with neuraminidase inhibitors, because these patients are not able to eradicate the virus from the respiratory tract, leading to the emergence of drug-resistant mutant viruses. Methods In this study, we examined the efficacy of baloxavir marboxil in nude mice that were immunologically deficient. Results Daily treatment with a suboptimal dose of baloxavir marboxil increased the survival time of the virus-infected nude mice but did not clear the virus from their respiratory organs, resulting in gradual body weight loss after termination of treatment. Conclusions Despite the prolonged baloxavir marboxil treatment, few resistant mutants were detected.
- Published
- 2019
11. Antibody-Dependent Enhancement of SARS-CoV-2 Infection Is Mediated by the IgG Receptors FcγRIIA and FcγRIIIA but Does Not Contribute to Aberrant Cytokine Production by Macrophages
- Author
-
Seiya Yamayoshi, Makoto Kuroda, Peter Halfmann, Tammy Armbrust, Yoshihiro Kawaoka, and Tadashi Maemura
- Subjects
Chemokine ,Feline coronavirus ,viruses ,medicine.medical_treatment ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,Microbiology ,Virus ,Cell Line ,Proinflammatory cytokine ,Immune system ,Cricetinae ,Virology ,medicine ,Animals ,Humans ,Antibody-dependent enhancement ,FcγRIIA ,biology ,SARS-CoV-2 ,Macrophages ,Receptors, IgG ,COVID-19 ,Antibody-Dependent Enhancement ,QR1-502 ,FcγRIIIA ,Cytokine ,Immunology ,biology.protein ,Cytokines ,Antibody ,ADE ,Research Article - Abstract
The coronavirus disease 2019 (COVID-19) pandemic has raised concerns about the detrimental effects of antibodies. Antibody-dependent enhancement (ADE) of infection is one of the biggest concerns in terms of not only the antibody reaction to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) upon reinfection with the virus but also the reaction to COVID-19 vaccines. In this study, we evaluated ADE of infection by using COVID-19 convalescent-phase plasma and BHK cells expressing human Fcγ receptors (FcγRs). We found that FcγRIIA and FcγRIIIA mediated modest ADE of infection against SARS-CoV-2. Although ADE of infection was observed in monocyte-derived macrophages infected with SARS-CoV-2, including its variants, proinflammatory cytokine/chemokine expression was not upregulated in macrophages. SARS-CoV-2 infection thus produces antibodies that elicit ADE of infection, but these antibodies do not contribute to excess cytokine production by macrophages. IMPORTANCE Viruses infect cells mainly via specific receptors at the cell surface. Antibody-dependent enhancement (ADE) of infection is an alternative mechanism of infection for viruses to infect immune cells that is mediated by antibodies and IgG receptors (FcγRs). Because ADE of infection contributes to the pathogenesis of some viruses, such as dengue virus and feline coronavirus, it is important to evaluate the precise mechanism of ADE and its contribution to the pathogenesis of SARS-CoV-2. Here, using convalescent-phase plasma from COVID-19 patients, we found that two types of FcγRs, FcγRIIA and FcγRIIIA, mediate ADE of SARS-CoV-2 infection. Although ADE of infection was observed for SARS-CoV-2 and its recent variants, proinflammatory cytokine production in monocyte-derived macrophages was not upregulated. These observations suggest that SARS-CoV-2 infection produces antibodies that elicit ADE of infection, but these antibodies may not be involved in aberrant cytokine release by macrophages during SARS-CoV-2 infection.
- Published
- 2021
12. SARS-CoV-2 spike P681R mutation enhances and accelerates viral fusion
- Author
-
Masaki Imai, Isao Yoshida, Takashi Irie, Kiyoko Iwatsuki-Horimoto, Seiya Ozono, Ryoko Kawabata, Takasuke Fukuhara, So Nakagawa, Seiya Yamayoshi, Mami Nagashima, Yoshihito Horisawa, Yuri L Tanaka, Terumasa Ikeda, Rigel Suzuki, Erika P Butlertanaka, Akifumi Takaori-Kondo, Ryosuke Nomura, Izumi Kimura, Takemasa Sakaguchi, Jiaqi Wu, Hesham Nasser, Mutsumi Ito, Ryo Shimizu, Jumpei Ito, Keiya Uriu, Kotaro Shirakawa, Yusuke Kosugi, Kazuhisa Yoshimura, Yasuhiro Kazuma, Kenzo Tokunaga, Akatsuki Saito, Kumiko Yoshimatsu, Kenji Sadamasu, Kenta Shimizu, Yoshihiro Kawaoka, Tadashi Maemura, Kei Sato, and Hiroyuki Asakura
- Subjects
Delta ,Mutation ,Lineage (genetic) ,medicine ,Spike (database) ,Biology ,Pathogenicity ,medicine.disease_cause ,Virology ,Genome ,Phenotype ,Virus - Abstract
SummaryDuring the current SARS-CoV-2 pandemic, a variety of mutations have been accumulated in the viral genome, and at least five variants of concerns (VOCs) have been considered as the hazardous SARS-CoV-2 variants to the human society. The newly emerging VOC, the B.1.617.2 lineage (delta variant), closely associates with a huge COVID-19 surge in India in Spring 2021. However, its virological property remains unclear. Here, we show that the B.1.617 variants are highly fusogenic and form prominent syncytia. Bioinformatic analyses reveal that the P681R mutation in the spike protein is highly conserved in this lineage. Although the P681R mutation decreases viral infectivity, this mutation confers the neutralizing antibody resistance. Notably, we demonstrate that the P681R mutation facilitates the furin-mediated spike cleavage and enhances and accelerates cell-cell fusion. Our data suggest that the P681R mutation is a hallmark characterizing the virological phenotype of this newest VOC, which may associate with viral pathogenicity.HighlightsP681R mutation is highly conserved in the B.1.617 lineagesP681R mutation accelerates and enhances SARS-CoV-2 S-mediated fusionPromotion of viral fusion by P681R mutation is augmented by TMPRSS2
- Published
- 2021
13. Uncovering the Anti-Ebola Repertome
- Author
-
Seiya Yamayoshi and Yoshihiro Kawaoka
- Subjects
Disease onset ,Proteome ,Disease ,Antibodies, Viral ,medicine.disease_cause ,Microbiology ,Article ,03 medical and health sciences ,0302 clinical medicine ,VP40 ,Antigen ,Virology ,medicine ,Humans ,Survivors ,030304 developmental biology ,Ebolavirus ,0303 health sciences ,biology ,Hemorrhagic Fever, Ebola ,humanities ,Antibody response ,biology.protein ,Parasitology ,Antibody ,030217 neurology & neurosurgery - Abstract
Evolution of antibody repertoire against the Ebola virus (EBOV) proteome was characterized in an acutely infected patient receiving supportive care alone to elucidate virus-host interactions over time. Differential kinetics are observed for IgM/IgG/IgA epitope diversity, antibody binding, and affinity maturation to EBOV proteins. During acute illness, antibodies predominate to VP40 and Glycoprotein (GP). At day 13 of clinical illness a marked increase in antibody titers to most EBOV proteins and affinity maturation to GP is associated with rapid decline in viral replication and illness severity. At one-year, despite undetectable virus, a diverse IgM repertoire against VP40 and GP epitopes is observed suggesting occult viral persistence. Rabbit immunization experiments identify key immunodominant sites of GP, while challenge studies in mice found these epitopes induce EBOV-neutralizing antibodies and protect against lethal EBOV challenge. This study reveals markers of viral persistence and provides promising approaches for development and evaluation of vaccines and therapeutics.
- Published
- 2020
14. Characterization of H7N9 avian influenza viruses isolated from duck meat products
- Author
-
Huapeng Feng, Maki Kiso, Yoshihiro Kawaoka, Akihiro Shibata, Hiromichi Mitake, Kiyoko Iwatsuki‐Hirimoto, Mutsumi Ito, Seiya Yamayoshi, Tiago J. S. Lopes, Hiroyuki Osaka, Tokiko Watanabe, Li Wu, Riho Sumiyoshi, and Masaki Imai
- Subjects
040301 veterinary sciences ,animal diseases ,viruses ,Highly pathogenic ,Chick Embryo ,Biology ,Influenza A Virus, H7N9 Subtype ,medicine.disease_cause ,Virus ,Madin Darby Canine Kidney Cells ,0403 veterinary science ,Mice ,03 medical and health sciences ,Dogs ,Japan ,Influenza, Human ,medicine ,Animals ,Humans ,Poultry Products ,Poultry Diseases ,030304 developmental biology ,Mice, Inbred BALB C ,0303 health sciences ,General Veterinary ,General Immunology and Microbiology ,Ferrets ,virus diseases ,04 agricultural and veterinary sciences ,General Medicine ,Pathogenicity ,Virology ,Low pathogenic ,Influenza A virus subtype H5N1 ,Ducks ,Influenza in Birds ,Female - Abstract
Avian influenza H7N9 viruses have caused five epidemic waves of human infections since the first human cases were reported in 2013. In 2016, the initial low pathogenic avian influenza (LPAI) H7N9 viruses became highly pathogenic, acquiring multi-basic amino acids at the haemagglutinin cleavage site. These highly pathogenic avian influenza (HPAI) H7N9 viruses have been detected in poultry and humans in China, causing concerns of a serious threat to global public health. In Japan, both HPAI and LPAI H7N9 viruses were isolated from duck meat products carried illegally and relinquished voluntarily at the border by passengers on flights from China to Japan between 2016 and 2017. Some of the LPAI and HPAI H7N9 viruses detected at the border in Japan were characterized previously in chickens and ducks; however, their pathogenicity and replicative ability in mammals remain unknown. In this study, we assessed the biological features of two HPAI H7N9 virus isolates [A/duck/Japan/AQ-HE29-22/2017 (HE29-22) and A/duck/Japan/AQ-HE29-52/2017 (HE29-52); both of these viruses were isolated from duck meat at the border)] and an LPAI H7N9 virus isolate [A/duck/Japan/AQ-HE28-3/2016 (HE28-3)] in mice and ferrets. In mice, HE29-52 was more pathogenic than HE29-22 and HE28-3. In ferrets, the two HPAI virus isolates replicated more efficiently in the lower respiratory tract of the animals than did the LPAI virus isolate. Our results indicate that HPAI H7N9 viruses with the potential to cause severe diseases in mammals have been illegally introduced to Japan.
- Published
- 2019
15. A single amino acid change in hemagglutinin reduces the cross-reactivity of antiserum against an equine influenza vaccine strain
- Author
-
Seiya Yamayoshi, Hiroshi Bannai, Takashi Yamanaka, Koji Tsujimura, Manabu Nemoto, Yoshihiro Kawaoka, and Hiroshi Kokado
- Subjects
Hemagglutinin (influenza) ,Hemagglutinin Glycoproteins, Influenza Virus ,Cross Reactions ,Biology ,Antibodies, Viral ,medicine.disease_cause ,Cross-reactivity ,03 medical and health sciences ,Orthomyxoviridae Infections ,Virology ,medicine ,Animals ,Horses ,Pathogen ,030304 developmental biology ,Antiserum ,0303 health sciences ,030306 microbiology ,Immune Sera ,Antibody titer ,General Medicine ,Reverse genetics ,Vaccination ,Amino Acid Substitution ,Influenza A virus ,Influenza Vaccines ,biology.protein ,Horse Diseases ,Antibody - Abstract
Equine influenza virus is an important pathogen for the horse industry because of its economic impact, and vaccination is a key control measure. Our previous work suggested that a mutation at position 144 in the hemagglutinin of Florida sublineage clade 2 viruses reduces the cross-neutralizing activity of antiserum against a former vaccine strain. To confirm this suggestion, here, we generated viruses by reverse genetics. Antibody titers against the mutated viruses were one-tenth to one-sixteenth of those against the former vaccine strain. Our findings confirm that this single amino acid substitution reduces the cross-reactivity of antiserum against this former Japanese vaccine.
- Published
- 2019
16. Antibody-free digital influenza virus counting based on neuraminidase activity
- Author
-
Yoichiro Fujioka, Seiya Yamayoshi, Kazuhito V. Tabata, Yusuke Ohba, Yoshihiro Kawaoka, Yoshiki Moriizumi, Yoshihiro Minagawa, Yuko Kawaguchi, Hiroyuki Noji, and Mana Ono
- Subjects
0301 basic medicine ,Oseltamivir ,viruses ,Neuraminidase ,lcsh:Medicine ,medicine.disease_cause ,Virus ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Viral Proteins ,0302 clinical medicine ,Influenza A Virus, H1N1 Subtype ,Influenza A virus ,medicine ,lcsh:Science ,Detection limit ,Multidisciplinary ,biology ,Chemistry ,Influenza A Virus, H3N2 Subtype ,lcsh:R ,Virion ,Virology ,Titer ,030104 developmental biology ,biology.protein ,Alkaline phosphatase ,lcsh:Q ,Antibody ,030217 neurology & neurosurgery - Abstract
There is large demand for a quantitative method for rapid and ultra-sensitive detection of the influenza virus. Here, we established a digital influenza virus counting (DIViC) method that can detect a single virion without antibody. In the assay, a virion is stochastically entrapped inside a femtoliter reactor array device for the fluorogenic assay of neuraminidase, and incubated for minutes. By analyzing 600,000 reactors, the practical limit of detection reached the order of 103 (PFU)/mL, only 10-times less sensitive than RT-PCR and more than 1000-times sensitive than commercial rapid test kits (RIDTs). Interestingly, neuraminidase activity differed among virions. The coefficient of variance was 30–40%, evidently broader than that of alkaline phosphatase measured as a model enzyme for comparison, suggesting the heterogeneity in size and integrity among influenza virus particles. Sensitivity to oseltamivir also differed between virions. We also tested DIViC using clinical gargle samples that imposes less burden for sampling while with less virus titre. The comparison with RIDTs showed that DIViC was largely superior to RIDTs in the sensitivity with the clinical samples although a few false-positive signals were observed in some clinical samples that remains as a technical challenge.
- Published
- 2019
17. Optimization of an LNP-mRNA vaccine candidate targeting SARS-CoV-2 receptor-binding domain
- Author
-
Yasushi Itoh, Kiyoko Iwatsuki-Horimoto, Mutsumi Ito, Nao Jounai, Hioki Kou, Seiya Yamayoshi, Yoshinori Kitagawa, Takako Niwa, Fumihiko Takeshita, Takashi Suzuki, Yoshihiro Kawaoka, Misako Nakayama, Kouji Kobiyama, Ken Ishii, Shintaro Yamada, Temizoz B, Negishi H, Tsuchida J, Masayuki Imai, Cong Thanh Nguyen, Tomoyuki Watanabe, Maki Kiso, and Hirohito Ishigaki
- Subjects
Messenger RNA ,Reactogenicity ,Innate immune system ,Immunization ,Interferon ,Immunogenicity ,medicine ,Biology ,medicine.disease_cause ,Type I interferon production ,Virology ,Coronavirus ,medicine.drug - Abstract
In 2020, two mRNA-based vaccines, encoding the full length of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein, have been introduced for control of the coronavirus disease (COVID-19) pandemic1,2. However, reactogenicity, such as fever, caused by innate immune responses to the vaccine formulation remains to be improved. Here, we optimized a lipid nanoparticle (LNP)-based mRNA vaccine candidate, encoding the SARS-CoV-2 spike protein receptor-binding domain (LNP-mRNA-RBD), which showed improved immunogenicity by removing reactogenic materials from the vaccine formulation and protective potential against SARS-CoV-2 infection in cynomolgus macaques. LNP-mRNA-RBD induced robust antigen-specific B cells and follicular helper T cells in the BALB/c strain but not in the C57BL/6 strain; the two strains have contrasting abilities to induce type I interferon production by dendritic cells. Removal of reactogenic materials from original synthesized mRNA by HPLC reduced type I interferon (IFN) production by dendritic cells, which improved immunogenicity. Immunization of cynomolgus macaques with an LNP encapsulating HPLC-purified mRNA induced robust anti-RBD IgG in the plasma and in various mucosal areas, including airways, thereby conferring protection against SARS-CoV-2 infection. Therefore, fine-tuning the balance between the immunogenic and reactogenic activity of mRNA-based vaccine formulations may offer safer and more efficacious outcomes.
- Published
- 2021
18. Chimeric hPIV2/Corona-Spike Nasal Vaccine Robustly Protects the Upper and Lower Airways Against SARS-CoV-2
- Author
-
Ryoichi Ono, Seiya Yamayoshi, Yoshihiro Kawaoka, Tadashi Maemura, Masayuki Fukumura, Junpei Ohtsuka, Asami Eguchi, Maeda M, Mutsumi Ito, Yosky Kataoka, Tetsuya Nosaka, and Masaki Imai
- Subjects
biology ,business.industry ,Virology ,Viral vector ,Herd immunity ,Vaccination ,Viral envelope ,Animal Care Committees ,biology.protein ,Medicine ,Nasal administration ,Vector (molecular biology) ,Antibody ,business - Abstract
We developed an intranasal vaccine against SARS-CoV-2 using the replication-incompetent human parainfluenza virus type 2 (hPIV2) vector BC-PIV, which can deliver ectopic gene as stable RNA and ectopic protein on the envelope. BC-PIV expressing the full-length prefusion-stabilized spike gene of SARS-CoV-2, S2PM, possessed a corona-like viral envelope. Intranasal vaccination of mice with BC-PIV/S2PM induced high levels of neutralizing IgG and mucosal IgA antibodies against the spike protein. While BC-PIV showed hemagglutinating activity, BC-PIV/S2PM lacked such activity, in accordance with the presence of the massive spike protein on the viral surface. Furthermore, single-dose intranasal vaccination of hamsters with BC-PIV/S2PM completely protected the lungs from SARS-CoV-2 at 11 weeks post-immunization, and prime-boost vaccination conferred virtually complete protection of the nasal turbinates against SARS-CoV-2 challenge at 11 weeks post-priming. Thus, this chimeric hPIV2/spike intranasal vaccine is one of the strong candidates for an ultimate vaccine against SARS-CoV-2 to curtail virus transmission. Funding: This work was supported in part by Grants-in-Aid from the Ministry of Education, Culture, Sports, Science and Technology in Japan (17K19652, 20K21614), by a Research Program on Emerging and Re-emerging Infectious Diseases from the Japan Agency for Medical Research and Development (AMED) (JP19fk0108113), Mie University (for research institutes of excellence), Mie Prefecture, Junior Chamber International Yokkaichi, and MediciNova, Inc. Conflict of Interest: J.O., M.F., M.Im., R.O., S.Y., Y.Kaw., and T.N. are patent applicants for recombinant BCPIV vaccine against SARS-CoV-2. M.F. is a founder of BioComo, Inc., and J.O. is an employee of BioComo, Inc. J.O., M.F., M.M., and T.N. have shares of stock in Biocomo, Inc. M.M. is a scientific advisor of JEOL Ltd. T.N. is a scientific advisor of MediciNova, Inc. The other authors declare no competing interests. Ethical Approval: Recombinant DNA experiments with SARS-CoV-2 S gene fragments were approved by the Ministry of Education, Culture, Sports, Science and Technology in Japan (Approved No. 2019-728, 729; 2020-362, 373, 948). The animal studies were approved by the Animal Care Committees of Mie University(Approved No. 23-33) and the Animal Experiment Committee of the Institute of Medical Science, the University of Tokyo (Approved No. PA19-75), and all methods were performed under institutional regulations of animal experiments in accordance with the current national guidelines. Animal experiments using SARS-CoV-2 S gene fragments orSARS-CoV-2 were also approved by the Ministry of Education, Culture, Sports, Science and Technology in Japan (Approved No. 2019-728, 729; 2020-362, 373, 2020-948).
- Published
- 2021
19. Comparison of Rapid Antigen Tests for COVID-19
- Author
-
Masaki Imai, Eisuke Adachi, Michiko Koga, Kazuhiko Ikeuchi, Seiya Yamayoshi, Takato Nakamoto, Atsuhiro Yasuhara, Shinya Yamada, Mutsumi Ito, Kenji Maeda, Shin ichiro Hattori, Rie Baba, Yuko Sakai-Tagawa, Kiyoko Iwatsuki-Horimoto, Hidefumi Koh, Yuri Furusawa, Takahiro Fukui, Hiroshi Yotsuyanagi, Makoto Saito, Kensuke Fujita, Hiroaki Mitsuya, Norio Omagari, Fumimaro Ito, Ichiro Nakachi, Hiroyuki Nagai, Yoshihiro Kawaoka, Michiko Ujie, Osamu Akasaka, Takayuki Ogura, and Kei Yamamoto
- Subjects
0301 basic medicine ,Coronavirus disease 2019 (COVID-19) ,diagnosis ,Point-of-Care Systems ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,030106 microbiology ,lcsh:QR1-502 ,Real-Time Polymerase Chain Reaction ,Sensitivity and Specificity ,Article ,Virus ,lcsh:Microbiology ,COVID-19 Serological Testing ,Specimen Handling ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Virology ,Humans ,Medicine ,030212 general & internal medicine ,rapid antigen test ,Antigens, Viral ,False Negative Reactions ,Immunoassay ,medicine.diagnostic_test ,business.industry ,SARS-CoV-2 ,COVID-19 ,Infectious Diseases ,Real-time polymerase chain reaction ,Rapid antigen test ,business - Abstract
Reverse transcription-quantitative PCR (RT-qPCR)-based tests are widely used to diagnose coronavirus disease 2019 (COVID-19). As a result that these tests cannot be done in local clinics where RT-qPCR testing capability is lacking, rapid antigen tests (RATs) for COVID-19 based on lateral flow immunoassays are used for rapid diagnosis. However, their sensitivity compared with each other and with RT-qPCR and infectious virus isolation has not been examined. Here, we compared the sensitivity among four RATs by using severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) isolates and several types of COVID-19 patient specimens and compared their sensitivity with that of RT-qPCR and infectious virus isolation. Although the RATs read the samples containing large amounts of virus as positive, even the most sensitive RAT read the samples containing small amounts of virus as negative. Moreover, all RATs tested failed to detect viral antigens in several specimens from which the virus was isolated. The current RATs will likely miss some COVID-19 patients who are shedding infectious SARS-CoV-2.
- Published
- 2020
- Full Text
- View/download PDF
20. Antibody titers against SARS-CoV-2 decline, but do not disappear for several months
- Author
-
Hiroaki Mitsuya, Takayuki Ogura, Shiho Chiba, Jurika Murakami, Michiko Koga, Junichi Ochi, Masanori Nojima, Ichiro Nakachi, Tetsuya Suzuki, Hiroyuki Nagai, Kenji Maeda, Norio Omagari, Kensuke Fujita, Makoto Saito, Osamu Akasaka, Rie Baba, Hideaki Nakajima, Yoshihiro Kawaoka, Hideaki Kato, Kazuma Yagi, Norio Sugaya, Kiyoko Iwatsuki-Horimoto, Eisuke Adachi, David J. Pattinson, Taiki Hamabata, Mutsumi Ito, Yusuke Miyazato, Keiko Mitamura, Hiroshi Yotsuyanagi, Shin ichiro Hattori, Atsuhiro Yasuhara, Morio Nakamura, Shinya Yamamoto, Moe Okuda, and Seiya Yamayoshi
- Subjects
lcsh:R5-920 ,Allergy ,biology ,business.industry ,010102 general mathematics ,Antibody titer ,General Medicine ,medicine.disease ,01 natural sciences ,03 medical and health sciences ,Titer ,0302 clinical medicine ,Ectodomain ,Immunology ,Cohort ,biology.protein ,Medicine ,030212 general & internal medicine ,0101 mathematics ,Antibody ,lcsh:Medicine (General) ,business ,Neutralizing antibody ,Pathogen ,Research Paper - Abstract
Background To develop an effective vaccine against a novel viral pathogen, it is important to understand the longitudinal antibody responses against its first infection. Here we performed a longitudinal study of antibody responses against SARS-CoV-2 in symptomatic patients. Methods Sequential blood samples were collected from 39 individuals at various timepoints between 0 and 154 days after onset. IgG or IgM titers to the receptor binding domain (RBD) of the S protein, the ectodomain of the S protein, and the N protein were determined by using an ELISA. Neutralizing antibody titers were measured by using a plaque reduction assay. Findings The IgG titers to the RBD of the S protein, the ectodomain of the S protein, and the N protein peaked at about 20 days after onset, gradually decreased thereafter, and were maintained for several months after onset. Extrapolation modeling analysis suggested that the IgG antibodies were maintained for this amount of time because the rate of reduction slowed after 30 days post-onset. IgM titers to the RBD decreased rapidly and disappeared in some individuals after 90 days post-onset. All patients, except one, possessed neutralizing antibodies against authentic SARS-CoV-2, which they retained at 90 days after onset. The highest antibody titers in patients with severe infections were higher than those in patients with mild or moderate infections, but the decrease in antibody titer in the severe infection cohort was more remarkable than that in the mild or moderate infection cohort. Interpretation Although the number of patients is limited, our results show that the antibody response against the first SARS-CoV-2 infection in symptomatic patients is typical of that observed in an acute viral infection. Funding The Japan Agency for Medical Research and Development and the National Institutes of Allergy and Infectious Diseases.
- Published
- 2020
21. Longitudinal antibody repertoire in 'mild' versus 'severe' COVID-19 patients reveals immune markers associated with disease severity and resolution
- Author
-
Makoto Saito, Surender Khurana, Atsuhiro Yasuhara, Seiya Yamayoshi, Shinya Yamada, Takayuki Ogura, Laura Klenow, Masaki Imai, Osamu Akasaka, Kiyoko Iwatsuki-Horimoto, Eisuke Adachi, Elizabeth M. Coyle, Yuko Sakai-Tagawa, Youri Lee, Hiroshi Yotsuyanagi, Supriya Ravichandran, Rie Baba, Ichiro Nakachi, Mutsumi Ito, Maki Kiso, Gabrielle Grubbs, Michiko Koga, and Yoshihiro Kawaoka
- Subjects
Antibody Affinity ,macromolecular substances ,Antibodies, Viral ,Severity of Illness Index ,Epitope ,Affinity maturation ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antibody Repertoire ,Antigen ,Protein Domains ,Neutralization Tests ,Medicine ,Humans ,030212 general & internal medicine ,skin and connective tissue diseases ,Antigens, Viral ,Research Articles ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,biology ,business.industry ,SARS-CoV-2 ,fungi ,COVID-19 ,SciAdv r-articles ,biochemical phenomena, metabolism, and nutrition ,Viral Load ,Immunoglobulin Class Switching ,body regions ,Hospitalization ,Coronavirus ,Kinetics ,HEK293 Cells ,Immunoglobulin class switching ,Immunoglobulin M ,Immunology ,Antibody Formation ,Spike Glycoprotein, Coronavirus ,biology.protein ,Cytokines ,Antibody ,business ,Biomarkers ,Protein Binding ,Research Article - Abstract
SARS-CoV-2–induced human antibody repertoire in patients with COVID-19 reveals immune markers of disease severity and recovery., Limited knowledge exists on immune markers associated with disease severity or recovery in patients with coronavirus disease 2019 (COVID-19). Here, we elucidated longitudinal evolution of SARS-CoV-2 antibody repertoire in patients with acute COVID-19. Differential kinetics was observed for immunoglobulin M (IgM)/IgG/IgA epitope diversity, antibody binding, and affinity maturation in “severe” versus “mild” COVID-19 patients. IgG profile demonstrated immunodominant antigenic sequences encompassing fusion peptide and receptor binding domain (RBD) in patients with mild COVID-19 who recovered early compared with “fatal” COVID-19 patients. In patients with severe COVID-19, high-titer IgA were observed, primarily against RBD, especially in patients who succumbed to SARS-CoV-2 infection. The patients with mild COVID-19 showed marked increase in antibody affinity maturation to prefusion SARS-CoV-2 spike that associated with faster recovery from COVID-19. This study revealed antibody markers associated with disease severity and resolution of clinical disease that could inform development and evaluation of effective immune-based countermeasures against COVID-19.
- Published
- 2020
22. Syrian hamsters as a small animal model for SARS-CoV-2 infection and countermeasure development
- Author
-
Yuri Furusawa, Noriko Kinoshita, Noriko Nakajima, Tadaki Suzuki, Aaron Balogh, Tokiko Watanabe, Peter Halfmann, Samantha Loeber, Moe Okuda, Mutsumi Ito, Seiya Yamayoshi, Makoto Takeda, Lizheng Guan, Florian Krammer, Shufang Fan, Kenta Takahashi, Kiyoko Iwatsuki-Horimoto, Masaki Imai, Hiroshi Ueki, Tammy Armbrust, Masato Hatta, Shin-ichiro Hattori, Tiago J. S. Lopes, Atsuhiro Yasuhara, Shiho Chiba, Yuko Sakai-Tagawa, Norio Ohmagari, Shinya Yamada, Hiroaki Mitsuya, Makoto Kuroda, Michiko Ujie, Yoshihiro Kawaoka, Maki Kiso, and Kosuke Takada
- Subjects
0301 basic medicine ,viruses ,medicine.disease_cause ,Antibodies, Viral ,Virus Replication ,Pathogenesis ,0302 clinical medicine ,Cricetinae ,Chlorocebus aethiops ,Neutralizing antibody ,skin and connective tissue diseases ,Lung ,Coronavirus ,Multidisciplinary ,biology ,virus diseases ,respiratory system ,Biological Sciences ,medicine.anatomical_structure ,Ribonucleoproteins ,Coronavirus Infections ,Pneumonia, Viral ,Hamster ,Lung injury ,Microbiology ,Virus ,Cell Line ,03 medical and health sciences ,Betacoronavirus ,Viral Proteins ,medicine ,Animals ,Humans ,Pandemics ,Vero Cells ,COVID-19 Serotherapy ,Mesocricetus ,business.industry ,SARS-CoV-2 ,fungi ,Immunization, Passive ,COVID-19 ,biology.organism_classification ,Virology ,Antibodies, Neutralizing ,infection ,respiratory tract diseases ,Disease Models, Animal ,030104 developmental biology ,biology.protein ,business ,Syrian hamsters ,countermeasure ,030217 neurology & neurosurgery - Abstract
Significance Since SARS-CoV-2 emerged in China, it has spread rapidly around the world. Effective vaccines and therapeutics for SARS-CoV-2−induced disease (coronavirus disease 2019;COVID-19) are urgently needed. We found that SARS-CoV-2 isolates replicate efficiently in the lungs of Syrian hamsters and cause severe pathological lesions in the lungs of these animals similar to commonly reported imaging features of COVID-19 patients with pneumonia. SARS-CoV-2−infected hamsters mounted neutralizing antibody responses and were protected against rechallenge with SARS-CoV-2. Moreover, passive transfer of convalescent serum to naïve hamsters inhibited virus replication in their lungs. Syrian hamsters are a useful small animal model for the evaluation of vaccines, immunotherapies, and antiviral drugs., At the end of 2019, a novel coronavirus (severe acute respiratory syndrome coronavirus 2; SARS-CoV-2) was detected in Wuhan, China, that spread rapidly around the world, with severe consequences for human health and the global economy. Here, we assessed the replicative ability and pathogenesis of SARS-CoV-2 isolates in Syrian hamsters. SARS-CoV-2 isolates replicated efficiently in the lungs of hamsters, causing severe pathological lung lesions following intranasal infection. In addition, microcomputed tomographic imaging revealed severe lung injury that shared characteristics with SARS-CoV-2−infected human lung, including severe, bilateral, peripherally distributed, multilobular ground glass opacity, and regions of lung consolidation. SARS-CoV-2−infected hamsters mounted neutralizing antibody responses and were protected against subsequent rechallenge with SARS-CoV-2. Moreover, passive transfer of convalescent serum to naïve hamsters efficiently suppressed the replication of the virus in the lungs even when the serum was administrated 2 d postinfection of the serum-treated hamsters. Collectively, these findings demonstrate that this Syrian hamster model will be useful for understanding SARS-CoV-2 pathogenesis and testing vaccines and antiviral drugs.
- Published
- 2020
23. Rapid and accurate detection of novel coronavirus SARS-CoV-2 using CRISPR-Cas3
- Author
-
Hiroshi Yotsuyanagi, Masayuki Yamamoto, Satomi Shibumura, Kohei Takeshita, Seiya Yamayoshi, Yuko Yamauchi, Yoshihiro Kawaoka, Tomoji Mashimo, and Kazuto Yoshimi
- Subjects
Dna cleavage ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,medicine ,CRISPR ,In patient ,Severe acute respiratory syndrome coronavirus ,Biology ,medicine.disease_cause ,Diagnostic tools ,Virology ,Virus ,Coronavirus - Abstract
Novel coronavirus SARS-CoV-2 outbreaks have rapidly spread to multiple countries, highlighting the urgent necessity for fast, sensitive, and specific diagnostic tools for virus surveillance. Here, the previously unknown collateral single-stranded DNA cleavage we observed with type I CRISPR-Cas3 highlights its potential for development as a Cas3-mediated rapid (within 40 min), low-cost, instrument-free detection method for SARS-CoV-2. This Cas3-based assay is comparable with Cas12- and real-time reverse-transcriptase PCR-based assays in its speed and sensitivity, but offers greater specificity for single-base-pair discrimination while negating the need for highly trained operators. These findings support the use of CRISPR diagnostics for point-of-care testing in patients with suspected SARS-CoV-2 infections.
- Published
- 2020
24. Triple combination therapy of favipiravir plus two monoclonal antibodies eradicates influenza virus from nude mice
- Author
-
Yoshihiro Kawaoka, Seiya Yamayoshi, and Maki Kiso
- Subjects
0301 basic medicine ,Combination therapy ,medicine.drug_class ,viruses ,030106 microbiology ,Hemagglutinins, Viral ,Mice, Nude ,Medicine (miscellaneous) ,macromolecular substances ,Favipiravir ,Monoclonal antibody ,Antiviral Agents ,Article ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Mice ,03 medical and health sciences ,Influenza A Virus, H1N1 Subtype ,Immune system ,Orthomyxoviridae Infections ,Triple combination ,Viral neuraminidase ,Animals ,Medicine ,lcsh:QH301-705.5 ,business.industry ,Antibodies, Monoclonal ,Antivirals ,Amides ,Virology ,030104 developmental biology ,lcsh:Biology (General) ,Viral replication ,Pyrazines ,Drug Therapy, Combination ,Female ,Influenza virus ,General Agricultural and Biological Sciences ,business - Abstract
Prolonged treatment of immunocompromised influenza patients with viral neuraminidase (NA) inhibitors is required, because the immune system of such patients fails to eradicate the viruses. Here, we attempted to eradicate influenza virus from the respiratory organs of nude mice, which is a model of immunocompromised hosts, by using combination therapy of the viral polymerase inhibitor favipiravir and monoclonal antibodies (mAbs) against the receptor-binding site (RBS) and stem of viral hemagglutinin (HA). Although monotherapy or combination therapy of two antivirals (two mAbs or favipiravir plus a mAb) suppressed virus replication, they failed to eradicate viruses from nude mice. In contrast, the triple combination therapy of favipiravir plus anti-Stem and anti-RBS mAbs completely stopped virus replication in nude mice, resulting in virus clearance. Triple combination approaches should be considered for the treatment of human immunocompromised patients with severe influenza.
- Published
- 2020
25. Gargle Lavage as a Safe and Sensitive Alternative to Swab Samples to Diagnose COVID-19: A Case Report in Japan
- Author
-
Seiya Yamayoshi, Yoshihiro Kawaoka, Michiko Koga, Kiyoko Iwatsuki-Horimoto, Makoto Saito, Eisuke Adachi, and Hiroshi Yotsuyanagi
- Subjects
Microbiology (medical) ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,biology ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Therapeutic irrigation ,biology.organism_classification ,medicine.disease ,Virology ,Pneumonia ,AcademicSubjects/MED00290 ,Infectious Diseases ,Pandemic ,Correspondence ,Medicine ,business ,Coronavirus Infections ,Betacoronavirus - Published
- 2020
- Full Text
- View/download PDF
26. Rapid and Accurate Detection of Novel Coronavirus SARS-CoV-2 Using CRISPR-Cas3
- Author
-
Yoshihiro Kawaoka, Kazuto Yoshimi, Yuko Yamauchi, Hiroshi Yotsuyanagi, Kohei Takeshita, Seiya Yamayoshi, Tomoji Mashimo, Masayuki Yamamoto, and Satomi Shibumura
- Subjects
Science research ,Dna cleavage ,business.industry ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,medicine ,CRISPR ,Outbreak ,In patient ,Severe acute respiratory syndrome coronavirus ,Computational biology ,medicine.disease_cause ,business ,Coronavirus - Abstract
Novel coronavirus SARS-CoV-2 outbreaks have rapidly spread to multiple countries, highlighting the urgent necessity for fast, sensitive, and specific diagnostic tools for virus surveillance. Here, the previously unknown collateral single-stranded DNA cleavage we observed with type I CRISPR-Cas3 highlights its potential for development as a Cas3-mediated rapid (within 40 min), low-cost, instrument-free detection method for SARS-CoV-2. This Cas3-based assay is comparable with Cas12- and real-time reverse-transcriptase PCR-based assays in its speed and sensitivity, but offers greater specificity for single-base-pair discrimination while negating the need for highly trained operators. These findings support the use of CRISPR diagnostics for point-of-care testing in patients with suspected SARS-CoV-2 infections. Funding: This project was supported in part by JSPS KAKENHI Grant Number 18H03974 (T.M.) and 19K16025 (K.Y.). Partially support also came from the Platform Project for Supporting Drug Discovery and Life Science Research (Basis for Supporting Innovative Drug Discovery and Life Science Research (BINDS)) from AMED under Grant Number JP20am0101070 (support numbers 1251 and 2463), from Research Program on Emerging and Reemerging Infectious Diseases from AMED (JP19fk0108113), and from the National Institutes of Allergy and Infectious Diseases funded Center for Research on Influenza Pathogenesis (CRIP; HHSN272201400008C). Conflict of Interest: S.S. is employee of C4U. K.Y. and T.M. are co-founders and scientific advisors for C4U. The other authors declare no competing interests.
- Published
- 2020
27. N-Terminal Acetylation by NatB Is Required for the Shutoff Activity of Influenza A Virus PA-X
- Author
-
Seiya Yamayoshi, Yoshihiro Kawaoka, Hiroko Kozuka-Hata, Kohei Oishi, and Masaaki Oyama
- Subjects
0301 basic medicine ,Saccharomyces cerevisiae Proteins ,viruses ,Mutant ,Saccharomyces cerevisiae ,Viral Nonstructural Proteins ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Virus ,Article ,03 medical and health sciences ,Acetyltransferases ,Influenza A virus ,medicine ,Humans ,Polymerase ,030102 biochemistry & molecular biology ,biology ,Chemistry ,Acetylation ,Cell biology ,Repressor Proteins ,030104 developmental biology ,HEK293 Cells ,Posttranslational modification ,biology.protein ,Function (biology) - Abstract
Summary N-terminal acetylation is a major posttranslational modification in eukaryotes catalyzed by N-terminal acetyltransferases (NATs), NatA through NatF. Although N-terminal acetylation modulates diverse protein functions, little is known about its roles in virus replication. We found that NatB, which comprises NAA20 and NAA25, is involved in the shutoff activity of influenza virus PA-X. The shutoff activity of PA-X was suppressed in NatB-deficient cells, and PA-X mutants that are not acetylated by NatB showed reduced shutoff activities. We also evaluated the importance of N-terminal acetylation of PA, because PA-X shares its N-terminal sequence with PA. Viral polymerase activity was reduced in NatB-deficient cells. Moreover, mutant PAs that are not acetylated by NatB lost their function in the viral polymerase complex. Taken together, our findings demonstrate that N-terminal acetylation is required for the shutoff activity of PA-X and for viral polymerase activity.
- Published
- 2018
- Full Text
- View/download PDF
28. Influenza A virus nucleoprotein is acetylated by histone acetyltransferases PCAF and GCN5
- Author
-
Shiori Takenaka, Takeshi Noda, Ayaka Saitoh, Rina Yoh, Seiya Yamayoshi, Masaki Shoji, Yoshihiro Kawaoka, Masahiro Nakano, Takashi Kuzuhara, Dai Hatakeyama, Naho Ohmi, Rina Nagano, Yumie Arakaki, Tsugunori Komatsu, and Aki Masuda
- Subjects
0301 basic medicine ,Lysine Acetyltransferases ,Transcription, Genetic ,Virus Replication ,medicine.disease_cause ,Biochemistry ,influenza virus ,Epigenesis, Genetic ,Tandem Mass Spectrometry ,Transcriptional regulation ,Influenza A virus ,p300-CBP Transcription Factors ,nucleoprotein ,Histone Acetyltransferases ,biology ,Chemistry ,Viral Core Proteins ,RNA-Binding Proteins ,Nucleocapsid Proteins ,influenza virus A ,Cell biology ,acetyltransferase ,Histone ,PCAF ,RNA, Viral ,RNA Interference ,viral protein ,histone acetylase ,Viral protein ,Blotting, Western ,Microbiology ,NP ,03 medical and health sciences ,medicine ,Humans ,Amino Acid Sequence ,Molecular Biology ,acetylation ,Lysine ,Epithelial Cells ,Cell Biology ,030104 developmental biology ,A549 Cells ,Acetylation ,HAT ,biology.protein ,Protein Processing, Post-Translational ,GCN5 ,Chromatography, Liquid - Abstract
Histone acetylation plays crucial roles in transcriptional regulation and chromatin organization. Viral RNA of the influenza virus interacts with its nucleoprotein (NP), whose function corresponds to that of eukaryotic histones. NP regulates viral replication and has been shown to undergo acetylation by the cAMP-response element (CRE)–binding protein (CBP) from the host. However, whether NP is the target of other host acetyltransferases is unknown. Here, we show that influenza virus NP undergoes acetylation by the two host acetyltransferases GCN5 and P300/CBP-associated factor (PCAF) and that this modification affects viral polymerase activities. Western blot analysis with anti–acetyl-lysine antibody on cultured A549 human lung adenocarcinoma epithelial cells infected with different influenza virus strains indicated acetylation of the viral NP. A series of biochemical analyses disclosed that the host lysine acetyltransferases GCN5 and PCAF acetylate NP in vitro. MS experiments identified three lysine residues as acetylation targets in the host cells and suggested that Lys-31 and Lys-90 are acetylated by PCAF and GCN5, respectively. RNAi-mediated silencing of GCN5 and PCAF did not change acetylation levels of NP. However, interestingly, viral polymerase activities were increased by the PCAF silencing and were decreased by the GCN5 silencing, suggesting that acetylation of the Lys-31 and Lys-90 residues has opposing effects on viral replication. Our findings suggest that epigenetic control of NP via acetylation by host acetyltransferases contributes to regulation of polymerase activity in the influenza A virus.
- Published
- 2018
29. Enhanced Replication of Highly Pathogenic Influenza A(H7N9) Virus in Humans
- Author
-
Mutsumi Ito, Atsuhiro Yasuhara, Seiya Yamayoshi, Yuelong Shu, Maki Kiso, and Yoshihiro Kawaoka
- Subjects
0301 basic medicine ,Microbiology (medical) ,Genes, Viral ,Epidemiology ,Highly pathogenic ,030106 microbiology ,Virulence ,lcsh:Medicine ,medicine.disease_cause ,Influenza A Virus, H7N9 Subtype ,Virus Replication ,Virus ,lcsh:Infectious and parasitic diseases ,H7N9 ,03 medical and health sciences ,Mice ,respiratory infections ,Orthomyxoviridae Infections ,Replication (statistics) ,Influenza, Human ,medicine ,Animals ,Humans ,viruses ,lcsh:RC109-216 ,Polymerase ,Mutation ,biology ,lcsh:R ,Dispatch ,Virology ,030104 developmental biology ,Infectious Diseases ,Viral replication ,Amino Acid Substitution ,PB2 ,biology.protein ,influenza ,PA ,enhanced polymerase activity - Abstract
To clarify the threat posed by emergence of highly pathogenic influenza A(H7N9) virus infection among humans, we characterized the viral polymerase complex. Polymerase basic 2-482R, polymerase basic 2-588V, and polymerase acidic-497R individually or additively enhanced virus polymerase activity, indicating that multiple replication-enhancing mutations in 1 isolate may contribute to virulence.
- Published
- 2018
30. Human protective monoclonal antibodies against the HA stem of group 2 HAs derived from an H3N2 virus-infected human
- Author
-
Tadahiro Sasaki, Yoshihiro Kawaoka, Kazuyoshi Ikuta, Seiya Yamayoshi, Mutsumi Ito, and Ryuta Uraki
- Subjects
0301 basic medicine ,Microbiology (medical) ,medicine.drug_class ,030106 microbiology ,Hemagglutinin Glycoproteins, Influenza Virus ,Antibodies, Viral ,Monoclonal antibody ,medicine.disease_cause ,Article ,Germline ,Virus ,Epitope ,Epitopes ,03 medical and health sciences ,Neutralization Tests ,In vivo ,medicine ,Influenza A virus ,Animals ,Humans ,Mice, Inbred BALB C ,biology ,Influenza A Virus, H3N2 Subtype ,Antibodies, Monoclonal ,Antibodies, Neutralizing ,Virology ,In vitro ,HEK293 Cells ,030104 developmental biology ,Infectious Diseases ,Immunoglobulin G ,Mutation ,biology.protein ,Female ,Antibody - Abstract
Objectives Broadly reactive human monoclonal antibodies against the HA stem of influenza A virus are being developed as therapeutic agents as well as to understand the epitopes that are essential for a universal influenza virus vaccine. Methods We isolated and characterized two hetero-reactive human monoclonal antibodies from an H3N2 virus-infected human. Results These antibodies, which are predominantly bound to the HA stem of group 2 HAs, used IGHV3-66 and IGHV4-38-2 germline genes, respectively. They possessed in vitro neutralizing ability, and in vivo protective efficacy against lethal infection with H3N2 or H7N9 virus. Escape mutations revealed that one of the protective antibodies recognized the α-helix A of HA2, and the other recognized the C-terminal portion of the fusion peptide and the β-sheet that precedes the α-helix A of HA2. Conclusions Of many human protective monoclonal antibodies against the HA stem, two human protective monoclonal antibodies were isolated in this study that predominantly recognize epitopes on the HA stem of group 2 and use unique IGHV3-66 and IGHV4-38-2 germline genes.
- Published
- 2018
31. Functional analysis of Host proteins involved in RNA virus replication
- Author
-
Seiya Yamayoshi
- Subjects
viruses ,Active Transport, Cell Nucleus ,Biology ,Virus Replication ,medicine.disease_cause ,Virus ,Viral Matrix Proteins ,Mice ,VP40 ,medicine ,Animals ,Humans ,RNA Viruses ,COPII ,Ribonucleoprotein ,Receptors, Scavenger ,Ebola virus ,Viral matrix protein ,Lysosome-Associated Membrane Glycoproteins ,RNA-Binding Proteins ,RNA virus ,General Medicine ,biology.organism_classification ,Cell biology ,Viral replication ,Host-Pathogen Interactions ,COP-Coated Vesicles - Abstract
Since RNA virus genome encodes only a limited number of viral proteins, replication of RNA virus mostly relies on host cells. Elucidation of host proteins that play important roles in the virus replication cycles contributes not only to fundamental virology research but also to applied research such as development of antiviral drugs. We revealed that Ebola virus matrix protein VP40 utilized host COPII transport machinery for its intracellular transport to the plasma membrane. Second, we demonstrated that enterovirus A71 used Scavenger receptor class B member 2 (SCARB2) as a cellular receptor. Finally, we found that host protein CLUH played an important role in the subnuclear transport of influenza virus ribonucleoprotein (vRNP) complexes. Here, I would like to briefly introduce these findings.
- Published
- 2018
32. A Broadly Reactive Human Anti-hemagglutinin Stem Monoclonal Antibody That Inhibits Influenza A Virus Particle Release
- Author
-
Atsuhiro Yasuhara, Kohei Oishi, Mutsumi Ito, Kazuyoshi Ikuta, Sumiho Nakatsu, Ryuta Uraki, Maki Kiso, Seiya Yamayoshi, Tadahiro Sasaki, and Yoshihiro Kawaoka
- Subjects
0301 basic medicine ,medicine.drug_class ,030106 microbiology ,Antibody Affinity ,lcsh:Medicine ,Hemagglutinin (influenza) ,CHO Cells ,Virus Replication ,Monoclonal antibody ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Epitope ,Virus ,Madin Darby Canine Kidney Cells ,Epitopes ,Mice ,HA stem ,03 medical and health sciences ,Cricetulus ,Dogs ,Antigen ,Viral entry ,Cricetinae ,medicine ,Influenza A virus ,Animals ,Humans ,Cells, Cultured ,Virus Release ,lcsh:R5-920 ,biology ,lcsh:R ,Antibodies, Monoclonal ,General Medicine ,Human monoclonal antibody ,Virology ,HEK293 Cells ,Hemagglutinins ,030104 developmental biology ,Viral replication ,Broadly reactive ,biology.protein ,lcsh:Medicine (General) ,HeLa Cells ,Research Paper - Abstract
Many broadly reactive human monoclonal antibodies against the hemagglutinin (HA) stem of influenza A virus have been developed for therapeutic applications. These antibodies typically inhibit viral entry steps, especially the HA conformational change that is required for membrane fusion. To better understand the mechanisms by which such antibodies inhibit viral replication, we established broadly reactive human anti-HA stem antibodies and determined the properties of these antibodies by examining their reactivity with 18 subtypes of HA, evaluating their in vivo protective efficacy, identifying their epitopes, and characterizing their inhibitory mechanisms. Among the eight human monoclonal antibodies we generated, which recognized at least 3 subtypes of the soluble HA antigens tested, clone S9-1-10/5-1 reacted with 18 subtypes of HA and protected mice from lethal infection with H1N1pdm09, H3N2, H5N1, and H7N9 viruses. This antibody recognized the HA2 helix A in the HA stem, and inhibited virus particle release from infected cells but did not block viral entry completely. These results show that broadly reactive human anti-HA stem antibodies can exhibit protective efficacy by inhibiting virus particle release. These findings expand our knowledge of the mechanisms by which broadly reactive stem-targeting antibodies inhibit viral replication and provide valuable information for universal vaccine development., Highlights • A broadly mouse-protective anti-HA stem antibody, S9-1-10/5-1, was isolated. • S9-1-10/5-1 mainly inhibited virus release rather than virus entry. • S9-1-10/5-1 tethers virions via crosslinking HA molecules between neighboring virions. Broadly reactive human monoclonal antibodies against the influenza HA stem have received attention because of their potential utility against multiple HA subtypes. Some of these antibodies inhibit virus entry and/or protect mice via antibody-dependent cellular cytotoxicity. Here, we identified a human monoclonal antibody that suppresses virus propagation in vitro and in vivo by primarily inhibiting virus particle release. This finding provides another inhibitory mechanism of action for the anti-HA stem antibodies, indicating that the anti-HA stem antibodies could be potent anti-virals due to their pluripotency.
- Published
- 2017
33. Treatment of Highly Pathogenic H7N9 Virus-Infected Mice with Baloxavir Marboxil
- Author
-
Seiya Yamayoshi, Yoshihiro Kawaoka, Yuri Furusawa, Maki Kiso, and Masaki Imai
- Subjects
Dibenzothiepins ,0301 basic medicine ,Oseltamivir ,Pyridines ,Pyridones ,Morpholines ,Highly pathogenic ,030106 microbiology ,lcsh:QR1-502 ,highly pathogenic ,Biology ,Influenza A Virus, H7N9 Subtype ,Virus Replication ,medicine.disease_cause ,baloxavir marboxil ,Antiviral Agents ,Virus ,lcsh:Microbiology ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Orthomyxoviridae Infections ,Virology ,Oxazines ,medicine ,Viral neuraminidase ,Animals ,Respiratory system ,h7n9 ,Dose-Response Relationship, Drug ,Virulence ,Triazines ,Communication ,Viral Load ,Low pathogenic ,Influenza A virus subtype H5N1 ,030104 developmental biology ,Infectious Diseases ,Viral replication ,chemistry ,Thiepins ,influenza - Abstract
Viral neuraminidase inhibitors show limited efficacy in mice infected with H7N9 influenza A viruses isolated from humans. Although baloxavir marboxil protected mice from lethal challenge infection with a low pathogenic avian influenza H7N9 virus isolated from a human, its efficacy in mice infected with a recent highly pathogenic version of H7N9 human isolates is unknown. Here, we examined the efficacy of baloxavir marboxil in mice infected with a highly pathogenic human H7N9 virus, A/Guangdong/17SF003/2016. Treatment of infected mice with a single 1.5 mg/kg dose of baloxavir marboxil protected mice from the highly pathogenic human H7N9 virus infection as effectively as oseltamivir treatment at 50 mg/kg twice a day for five days. Daily treatment for five days at 15 or 50 mg/kg of baloxavir marboxil showed superior therapeutic efficacy, largely preventing virus replication in respiratory organs. These results indicate that baloxavir marboxil is a valuable candidate treatment for human patients suffering from highly pathogenic H7N9 virus infection.
- Published
- 2019
34. Sensitivity of Commercially Available Influenza Rapid Diagnostic Tests in the 2018–2019 Influenza Season
- Author
-
Yuko Sakai-Tagawa, Seiya Yamayoshi, and Yoshihiro Kawaoka
- Subjects
Microbiology (medical) ,Point-of-care testing ,viruses ,lcsh:QR1-502 ,specificity ,Influenza season ,Biology ,medicine.disease_cause ,Microbiology ,lcsh:Microbiology ,Virus ,Seasonal influenza ,rapid diagnosis test ,03 medical and health sciences ,medicine ,030304 developmental biology ,Original Research ,0303 health sciences ,030306 microbiology ,Diagnostic test ,virus diseases ,Influenza a ,sensitivity ,Virology ,Influenza A virus subtype H5N1 ,point-of-care testing ,influenza - Abstract
Epidemics of seasonal influenza caused by H1N1pdm09, H3N2, and type B viruses occur throughout the world. Sporadic human H5 and H7N9 virus infections are also reported in particular regions. To treat influenza patients effectively with antivirals, sensitive and broad-reactive influenza rapid diagnostic tests (IRDTs) are required. Here, we tested the sensitivity of 23 IRDTs during the 2018–2019 influenza season for their ability to detect H1N1pdm09, H3N2, H5N1, H5N6, H7N9, and Victoria- and Yamagata-lineage type B viruses. All IRDTs detected all influenza A and B viruses tested but with different sensitivities. Several IRDTs detected the H5 and H7 viruses and the seasonal viruses with similar sensitivity. Such IRDTs might be useful for diagnosing patients infected with H5 and H7 viruses.
- Published
- 2019
35. Subclade 2.2.1-Specific Human Monoclonal Antibodies That Recognize an Epitope in Antigenic Site A of Influenza A(H5) Virus HA Detected between 2015 and 2018
- Author
-
Ryuta Uraki, Mutsumi Ito, Yoshihiro Kawaoka, Seiya Yamayoshi, Moe Okuda, and Taiki Hamabata
- Subjects
0301 basic medicine ,medicine.drug_class ,lcsh:QR1-502 ,Hemagglutinin Glycoproteins, Influenza Virus ,medicine.disease_cause ,Monoclonal antibody ,Antibodies, Viral ,H5-HA ,Virus ,Epitope ,lcsh:Microbiology ,Article ,03 medical and health sciences ,Epitopes ,0302 clinical medicine ,Antigen ,Neutralization Tests ,Virology ,Influenza A virus ,medicine ,Humans ,Amino Acids ,Immune Evasion ,Hemagglutination assay ,biology ,Influenza A Virus, H5N1 Subtype ,Antibodies, Monoclonal ,Subclade ,Hemagglutination Inhibition Tests ,Antibodies, Neutralizing ,030104 developmental biology ,Infectious Diseases ,Influenza Vaccines ,030220 oncology & carcinogenesis ,Mutation ,biology.protein ,human monoclonal antibody ,escape mutant virus ,Antibody ,Epitope Mapping - Abstract
Highly pathogenic avian H5 influenza viruses persist among poultry and wild birds throughout the world. They sometimes cause interspecies transmission between avian and mammalian hosts. H5 viruses possessing the HA of subclade 2.3.4.4, 2.3.2.1, 2.2.1, or 7.2 were detected between 2015 and 2018. To understand the neutralizing epitopes of H5-HA, we characterized 15 human monoclonal antibodies (mAbs) against the HA of H5 viruses, which were obtained from volunteers who received the H5N1 vaccine that contains a subclade 2.2.1 or 2.1.3.2 virus as an antigen. Twelve mAbs were specific for the HA of subclade 2.2.1, two mAbs were specific for the HA of subclade 2.1.3.2, and one mAb was specific for the HA of both. Of the 15 mAbs analyzed, nine, which were specific for the HA of subclade 2.2.1, and shared the VH and VL genes, possessed hemagglutination inhibition and neutralizing activities, whereas the others did not. A single amino acid substitution or insertion at positions 144&ndash, 147 in antigenic site A conferred resistance against these nine mAbs to the subclade 2.2.1 viruses. The amino acids at positions 144&ndash, 147 are highly conserved among subclade 2.2.1, but differ from those of other subclades. These results show that the neutralizing epitope including amino acids at positions 144&ndash, 147 is targeted by human antibodies, and plays a role in the antigenic difference between subclade 2.2.1 and other subclades.
- Published
- 2019
- Full Text
- View/download PDF
36. Identification of Amino Acid Residues in Influenza A Virus PA-X That Contribute to Enhanced Shutoff Activity
- Author
-
Kohei Oishi, Seiya Yamayoshi, and Yoshihiro Kawaoka
- Subjects
Microbiology (medical) ,mutagenesis analysis ,viruses ,lcsh:QR1-502 ,Mutagenesis (molecular biology technique) ,medicine.disease_cause ,Microbiology ,lcsh:Microbiology ,Virus ,Serine ,03 medical and health sciences ,Endonuclease ,Influenza A virus ,medicine ,influenza A virus ,Proline ,Original Research ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,shutoff activity ,biology ,030306 microbiology ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,3. Good health ,Amino acid ,PA-X ,chemistry ,Acetylation ,mRNA degradation ,biology.protein - Abstract
The influenza virus protein PA-X modulates the host immune responses and viral pathogenicity through suppression of host protein expression. The endonuclease active site in the N-terminal region, the basic amino acid cluster in the C-terminal PA-X-specific region, and N-terminal acetylation of PA-X by NatB are important for the shutoff activity of PA-X. Here, we focused on the shutoff activity of PA-X derived from the A/California/04/2009 and A/WSN/33 viruses because these two PA-X proteins differ in their shutoff activity. Mutagenesis analysis revealed that proline and serine at positions 28 and 65, respectively, play a central role in this difference. Furthermore, we found that P28 and S65 also affect the shutoff activity of PA-X derived from other influenza virus subtypes. These data demonstrate that P28 and S65 contribute to enhanced shutoff activity of PA-X.
- Published
- 2019
- Full Text
- View/download PDF
37. Characterization of Mouse Monoclonal Antibodies Against the HA of A(H7N9) Influenza Virus
- Author
-
Atsuo Motojima, Seiya Yamayoshi, Kenji Saito, Kazunari Nakaishi, Yoshihiro Kawaoka, Mutsumi Ito, and Kazushi Murakami
- Subjects
0301 basic medicine ,China ,medicine.drug_class ,030106 microbiology ,lcsh:QR1-502 ,HA ,Hemagglutinin (influenza) ,Hemagglutinin Glycoproteins, Influenza Virus ,Monoclonal antibody ,medicine.disease_cause ,Antibodies, Viral ,Influenza A Virus, H7N9 Subtype ,Virus ,Neutralization ,Epitope ,Antigenic drift ,Article ,lcsh:Microbiology ,H7N9 ,03 medical and health sciences ,Epitopes ,Mice ,Orthomyxoviridae Infections ,Virology ,medicine ,Influenza A virus ,Animals ,Antigenic change ,Mice, Inbred BALB C ,Hemagglutination assay ,biology ,Antibodies, Monoclonal ,mouse monoclonal antibody ,Hemagglutination Inhibition Tests ,Antibodies, Neutralizing ,030104 developmental biology ,Infectious Diseases ,Amino Acid Substitution ,biology.protein ,Female ,Chickens - Abstract
Many cases of human infection with the H7N9 virus have been detected in China since 2013. H7N9 viruses are maintained in chickens and are transmitted to humans at live bird markets. During circulation in birds, H7N9 viruses have accumulated amino acid substitutions in their hemagglutinin (HA), which resulted in an antigenically change in the recent H7N9 viruses. Here, we characterized 46 mouse monoclonal antibodies against the HA of the prototype strain. 16 H7-HA-specific monoclonal antibodies (mAbs) possessed hemagglutination inhibition (HI) and neutralization activities by recognizing the major antigenic site A, four other H7-HA-specific clones also showed HI and neutralizing activities via recognition of the major antigenic sites A and D, seven mAbs that reacted with several HA subtypes and possibly recognized the HA stem partially protected mice from lethal infection with prototype H7N9 virus, and the remaining 19 mAbs had neither HI nor neutralization activity. All human H7N9 viruses tested showed a similar neutralization sensitivity to the first group of 16 mAbs, whereas human H7N9 viruses isolated in 2016‒2017 were not neutralized by a second group of 4 mAbs. These results suggest that amino acid substitutions at the epitope of the second mAb group appear to be involved in the antigenic drift of the H7N9 viruses. Further analysis is required to fully understand the antigenic change in H7N9 viruses.
- Published
- 2019
- Full Text
- View/download PDF
38. Genetic and antigenic characterisation of influenza A(H3N2) viruses isolated in Yokohama during the 2016/17 and 2017/18 influenza seasons
- Author
-
Shinji Watanabe, Derek J. Smith, Kohei Shimizu, Miki Akimoto, Tomoko Momoki, Hiroki Ozawa, Yoshihiro Kawaoka, Shigeo Sugita, Hideka Miura, Shuzo Usuku, Seiya Yamayoshi, Takahiro Toyozawa, Kazuya Nakamura, Seiichiro Fujisaki, David J. Pattinson, Miwako Saikusa, Chiharu Kawakami, Ichiro Okubo, Atsuhiro Yasuhara, Smith, Derek [0000-0002-2393-1890], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Antigenicity ,glycosylation ,Epidemiology ,viruses ,viral infections ,Molecular Sequence Data ,030106 microbiology ,HA ,haemagglutinin ,Hemagglutinin Glycoproteins, Influenza Virus ,Biology ,medicine.disease_cause ,Sialidase ,influenza virus ,Virus ,03 medical and health sciences ,Japan ,Virology ,Influenza, Human ,medicine ,Antigenic variation ,Humans ,Epidemics ,Clade ,Gene ,Phylogeny ,Antiserum ,Reverse Transcriptase Polymerase Chain Reaction ,Research ,Influenza A Virus, H3N2 Subtype ,Public Health, Environmental and Occupational Health ,Genetic Variation ,virus diseases ,Sequence Analysis, DNA ,H3N2 ,Influenza A virus subtype H5N1 ,Hemagglutinins ,030104 developmental biology ,antigenicity ,surveillance ,RNA, Viral ,Seasons ,influenza - Abstract
Background: Influenza A(H3N2) virus rapidly evolves to evade human immune responses, resulting in changes in the antigenicity of haemagglutinin (HA). Therefore, continuous genetic and antigenic analyses of A(H3N2) virus are necessary to detect antigenic mutants as quickly as possible. Aim: We attempted to phylogenetically and antigenically capture the epidemic trend of A(H3N2) virus infection in Yokohama, Japan during the 2016/17 and 2017/18 influenza seasons. Methods: We determined the HA sequences of A(H3N2) viruses detected in Yokohama, Japan during the 2016/17 and 2017/18 influenza seasons to identify amino acid substitutions and the loss or gain of potential N-glycosylation sites in HA, both of which potentially affect the antigenicity of HA. We also examined the antigenicity of isolates using ferret antisera obtained from experimentally infected ferrets. Results: Influenza A(H3N2) viruses belonging to six clades (clades 3C.2A1, 3C.2A1a, 3C.2A1b, 3C.2A2, 3C.2A3 and 3C.2A4) were detected during the 2016/17 influenza season, whereas viruses belonging to two clades (clades 3C.2A1b and 3C.2A2) dominated during the 2017/18 influenza season. The isolates in clades 3C.2A1a and 3C.2A3 lost one N-linked glycosylation site in HA relative to other clades. Antigenic analysis revealed antigenic differences among clades, especially clade 3C.2A2 and 3C.2A4 viruses, which showed distinct antigenic differences from each other and from other clades in the antigenic map. Conclusion: Multiple clades, some of which differed antigenically from others, co-circulated in Yokohama, Japan during the 2016/17 and 2017/18 influenza seasons.
- Published
- 2019
39. Antigenic Change in Human Influenza A(H2N2) Viruses Detected by Using Human Plasma from Aged and Younger Adult Individuals
- Author
-
Yasumichi Arai, Yukimasa Matsuzawa, Yoshihiro Kawaoka, Kiyoko Iwatsuki-Horimoto, Seiya Yamayoshi, Yoshinori Nishimoto, Taiki Hamabata, Satoshi Fukuyama, Yukiko Abe, Yui Go, Ron A. M. Fouchier, Tokiko Watanabe, Masaki Imai, Moe Okuda, and Virology
- Subjects
Adult ,Male ,Antigenicity ,medicine.drug_class ,viruses ,lcsh:QR1-502 ,Biology ,Antibodies, Viral ,Monoclonal antibody ,medicine.disease_cause ,lcsh:Microbiology ,Article ,Neutralization ,Antigenic drift ,Influenza A Virus, H2N2 Subtype ,Young Adult ,Antigen ,Neutralization Tests ,Virology ,aged individuals ,Influenza, Human ,medicine ,Influenza A virus ,Humans ,influenza A virus ,Neutralizing antibody ,Phylogeny ,antigenic drift ,Aged, 80 and over ,H2N2 ,Middle Aged ,Titer ,Infectious Diseases ,biology.protein ,antigenic change ,Female - Abstract
Human influenza A(H2N2) viruses emerged in 1957 and were replaced by A(H3N2) viruses in 1968. The antigenicity of human H2N2 viruses has been tested by using ferret antisera or mouse and human monoclonal antibodies. Here, we examined the antigenicity of human H2N2 viruses by using human plasma samples obtained from 50 aged individuals who were born between 1928 and 1933 and from 33 younger adult individuals who were born after 1962. The aged individuals possessed higher neutralization titers against H2N2 viruses isolated in 1957 and 1963 than those against H2N2 viruses isolated in 1968, whereas the younger adults who were born between 1962 and 1968 possessed higher neutralization titers against H2N2 viruses isolated in 1963 than those against other H2N2 viruses. Antigenic cartography revealed the antigenic changes that occurred in human H2N2 viruses during circulation in humans for 11 years, as detected by ferret antisera. These results show that even though aged individuals were likely exposed to more recent H2N2 viruses that are antigenically distinct from the earlier H2N2 viruses, they did not possess high neutralizing antibody titers to the more recent viruses, suggesting immunological imprinting of these individuals with the first H2N2 viruses they encountered and that this immunological imprinting lasts for over 50 years.
- Published
- 2019
40. Identification of Novel Adjuvants for Ebola Virus-Like Particle Vaccine
- Author
-
Yoshihiro Kawaoka, Seiya Yamayoshi, Tiago J. S. Lopes, Makoto Yamashita, Tokiko Watanabe, Huapeng Feng, Sumiho Nakatsu, and Masaki Imai
- Subjects
0301 basic medicine ,viruses ,medicine.medical_treatment ,Immunology ,lcsh:Medicine ,Disease ,medicine.disease_cause ,Article ,virus-like particle ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Virus-like particle ,Drug Discovery ,Medicine ,Pharmacology (medical) ,030212 general & internal medicine ,Pharmacology ,Ebola virus ,Ebola vaccine ,business.industry ,lcsh:R ,virus diseases ,Outbreak ,Virology ,Clinical trial ,030104 developmental biology ,Infectious Diseases ,adjuvants ,business ,Adjuvant - Abstract
Ebola virus disease is a severe disease, often fatal, with a mortality rate of up to 90%. Presently, effective treatment and safe prevention options for Ebola virus disease are not available. Therefore, there is an urgent need to develop control measures to prevent or limit future Ebola virus outbreaks. Ebola virus protein-based virus-like particle (VLP) and inactivated whole virion vaccines have demonstrated efficacy in animal models, and the addition of appropriate adjuvants may provide additional benefits to these vaccines, including enhanced immune responses. In this study, we screened 24 compounds from injectable excipients approved for human use in Japan and identified six compounds that significantly enhanced the humoral response to Ebola VLP vaccine in a murine model. Our novel adjuvant candidates for Ebola VLP vaccine have already been demonstrated to be safe when administered intramuscularly or subcutaneously, and therefore, they are closer to clinical trials than adjuvants whose safety profiles are unknown.
- Published
- 2020
41. Isolation and Characterization of Human Monoclonal Antibodies That Recognize the Influenza A(H1N1)pdm09 Virus Hemagglutinin Receptor-Binding Site and Rarely Yield Escape Mutant Viruses
- Author
-
Atsuhiro Yasuhara, Seiya Yamayoshi, Mutsumi Ito, Maki Kiso, Shinya Yamada, and Yoshihiro Kawaoka
- Subjects
0301 basic medicine ,Microbiology (medical) ,medicine.drug_class ,viruses ,Mutant ,lcsh:QR1-502 ,HA ,Hemagglutinin (influenza) ,Receptor binding site ,medicine.disease_cause ,Monoclonal antibody ,Microbiology ,lcsh:Microbiology ,Virus ,antiviral agent ,03 medical and health sciences ,0302 clinical medicine ,Influenza A virus ,medicine ,influenza A virus ,Original Research ,biology ,Influenza a ,Virology ,030104 developmental biology ,biology.protein ,human monoclonal antibody ,escape mutant virus ,Antibody ,030217 neurology & neurosurgery - Abstract
The influenza A virus rapidly mutates to escape from antibodies. Here, we isolated and characterized three human monoclonal antibodies (mAbs) that neutralize A(H1N1)pdm09 viruses. Generation of escape mutant viruses suggested that these antibodies recognized conserved residues of the receptor-binding site (RBS) of hemagglutinin (HA) and that mutant viruses that escaped from these mAbs rarely appeared. Moreover, the escape mutant viruses grew significantly slower than wild-type virus, indicating their reduced fitness. These results indicate that these three human mAbs against the RBS of HA have the potential to be anti-influenza agents with a low propensity for the development of resistant viruses.
- Published
- 2018
42. Antigenic drift originating from changes to the lateral surface of the neuraminidase head of influenza A virus
- Author
-
Eisuke Adachi, Yoshihiro Kawaoka, Seiya Yamayoshi, I-Hsuan Wang, Shinya Yamada, Tadashi Kikuchi, Yuko Sakai-Tagawa, Atsuhiro Yasuhara, Michiko Koga, and Maki Kiso
- Subjects
Microbiology (medical) ,Antigenicity ,medicine.drug_class ,Entropy ,Immunology ,Neuraminidase ,Sialidase ,Monoclonal antibody ,medicine.disease_cause ,Antibodies, Viral ,Applied Microbiology and Biotechnology ,Microbiology ,Antiviral Agents ,Antigenic drift ,Virus ,03 medical and health sciences ,Epitopes ,Mice ,Viral Proteins ,Influenza A Virus, H1N1 Subtype ,Orthomyxoviridae Infections ,Sequence Analysis, Protein ,Genetics ,medicine ,Influenza A virus ,Animals ,Humans ,Antigens, Viral ,030304 developmental biology ,Immune Evasion ,0303 health sciences ,Mice, Inbred BALB C ,biology ,030306 microbiology ,Chemistry ,Receptors, IgG ,Immunity ,Antibodies, Monoclonal ,Cell Biology ,Molecular biology ,Disease Models, Animal ,HEK293 Cells ,Mutation ,biology.protein ,Female ,Antibody - Abstract
Influenza viruses possess two surface glycoproteins, haemagglutinin and neuraminidase (NA). Although haemagglutinin plays a major role as a protective antigen, immunity to NA also contributes to protection. The NA protein consists of a stalk and a head portion, the latter of which possesses enzymatic NA (or sialidase) activity. Like haemagglutinin, NA is under immune pressure, which leads to amino acid alterations and antigenic drift. Amino acid changes accumulate around the enzymatic active site, which is located at the top of the NA head. However, amino acid alterations also accumulate at the lateral surface of the NA head. The reason for this accumulation remains unknown. Here, we isolated seven anti-NA monoclonal antibodies (mAbs) from individuals infected with A(H1N1)pdm09 virus. We found that amino acid mutations on the lateral surface of the NA head abolished the binding of all of these mAbs. All seven mAbs activated Fcγ receptor (FcγR)-mediated signalling pathways in effector cells and five mAbs possessed NA inhibition activity, but the other two did not; however, all seven protected mice from lethal challenge infection through their NA inhibition activity and/or FcγR-mediated antiviral activity. Serological analysis of individuals infected with A(H1N1)pdm09 virus revealed that some possessed or acquired the anti-NA-lateral-surface antibodies following infection. We also found antigenic drift on the lateral surface of the NA head of isolates from 2009 and 2015. Our results demonstrate that anti-lateral-surface mAbs without NA inhibition activity can provide protection by activating FcγR-mediated antiviral activity and can drive antigenic drift at the lateral surface of the NA head. These findings have implications for NA antigenic characterization in that they demonstrate that traditional NA inhibition assays are inadequate to fully characterize NA antigenicity. The isolation and characterization of seven monoclonal antibodies that recognize the lateral surface of the influenza virus neuraminidase (NA) head reveal that, even in absence of NA-inhibiting activity, anti-NA antibodies can provide protection and drive antigenic drift at the lateral surface of the NA.
- Published
- 2018
43. Current and future influenza vaccines
- Author
-
Yoshihiro Kawaoka and Seiya Yamayoshi
- Subjects
0301 basic medicine ,Extramural ,business.industry ,Influenza vaccine ,viruses ,Early detection ,Influenza epidemics ,General Medicine ,Orthomyxoviridae ,Virology ,General Biochemistry, Genetics and Molecular Biology ,Virus ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Influenza Vaccines ,030220 oncology & carcinogenesis ,Pandemic ,Host-Pathogen Interactions ,Influenza, Human ,Medicine ,Humans ,business ,Pandemics - Abstract
Although antiviral drugs and vaccines have reduced the economic and healthcare burdens of influenza, influenza epidemics continue to take a toll. Over the past decade, research on influenza viruses has revealed a potential path to improvement. The clues have come from accumulated discoveries from basic and clinical studies. Now, virus surveillance allows researchers to monitor influenza virus epidemic trends and to accumulate virus sequences in public databases, which leads to better selection of candidate viruses for vaccines and early detection of drug-resistant viruses. Here we provide an overview of current vaccine options and describe efforts directed toward the development of next-generation vaccines. Finally, we propose a plan for the development of an optimal influenza vaccine.
- Published
- 2018
44. Differences in the ease with which mutant viruses escape from human monoclonal antibodies against the HA stem of influenza A virus
- Author
-
Ryuta Uraki, Atsuhiro Yasuhara, Yoshihiro Kawaoka, Mutsumi Ito, and Seiya Yamayoshi
- Subjects
0301 basic medicine ,medicine.drug_class ,030106 microbiology ,Mutant ,Hemagglutinin Glycoproteins, Influenza Virus ,medicine.disease_cause ,Monoclonal antibody ,Antibodies, Viral ,Virus ,Neutralization ,Epitope ,03 medical and health sciences ,Epitopes ,Mice ,Virology ,Influenza A virus ,medicine ,Animals ,Humans ,Immune Evasion ,Mice, Inbred BALB C ,biology ,Immunization, Passive ,Antibodies, Monoclonal ,Antibodies, Neutralizing ,030104 developmental biology ,Infectious Diseases ,Mutation ,biology.protein ,Female ,Antibody ,Clone (B-cell biology) - Abstract
Background Broadly protective human monoclonal antibodies that recognize the conserved epitopes in the HA of influenza A virus are being developed as therapeutic agents. Emergence of resistant viruses must always be considered when developing therapeutic agents against influenza. Objectives We examined human hetero-reactive mAbs against the HA stem of influenza A virus for the ease with which escape mutant viruses emerged. Study design We attempted to generate the mutant viruses escaped from the hetero-reactive anti-HA stem antibodies. We also evaluated their protective efficacy, binding affinity, and epitopes. Results We obtained several human monoclonal antibodies (mAbs) that react with the HA of different HA subtypes of influenza A virus belonging to group 1. Upon attempting to generate escape mutant viruses, we found that the ease with which such viruses emerged differed among the mAbs; viruses barely escaped from two of the mAbs (clones S9-3-37 and F20C77), whereas escape from the third mAb (clone F5B7) occurred readily. Comparisons of the mAbs revealed that the HA stem epitopes, in vitro neutralization potency, binding affinity to H1-HA, and protective efficacy against lethal challenge with H1N1pdm09 virus were all comparable. Conclusions These results demonstrate the importance of determining the ease with which escape mutant viruses emerge when evaluating anti-HA stem antibodies as antiviral agents during preclinical testing.
- Published
- 2018
45. Strain-Specific Contribution of Eukaryotic Elongation Factor 1 Gamma to the Translation of Influenza A Virus Proteins
- Author
-
Shuhei Sammaibashi, Seiya Yamayoshi, and Yoshihiro Kawaoka
- Subjects
0301 basic medicine ,Microbiology (medical) ,Protein subunit ,viruses ,lcsh:QR1-502 ,Biology ,medicine.disease_cause ,Microbiology ,lcsh:Microbiology ,Virus ,influenza virus ,eEF1G ,03 medical and health sciences ,0302 clinical medicine ,Influenza A virus ,medicine ,CRISPR ,Gene ,protein translation ,Original Research ,Messenger RNA ,Translation (biology) ,Cell biology ,030104 developmental biology ,Viral replication ,PB2 ,host protein ,030217 neurology & neurosurgery ,PA - Abstract
Influenza A virus exploits multiple host proteins during infection. To define the virus-host interactome, our group conducted a proteomics-based screen and identified 299 genes that contributed to virus replication and 24 genes that were antiviral. Of these genes, we focused on the role during virus replication of eukaryotic Elongation Factor 1 gamma (eEF1G), which is a subunit of the eukaryotic Elongation Factor-1 (eEF1) complex and known to be a pro-viral host protein. Using the CRISPR/Cas9 system, we obtained two clones that were defective in eEF1G expression. In both of these clones, A/WSN/33 (H1N1) virus growth and protein expression were significantly suppressed, but viral mRNA, vRNA, and cRNA expression were not reduced. However, the replication and protein expression of A/California/04/2009 (H1N1pdm) virus in both clones were similar to those in parental cells. We found that the PB2 and PA proteins of WSN virus were responsible for the eEF1G-dependent replication. Our data show that eEF1G plays a role in the translation of virus proteins in a strain-specific manner. Additional analyses may be needed to further understand the role of strain-specific host proteins during virus replication.
- Published
- 2018
46. Evaluation of the fusion partner cell line SPYMEG for obtaining human monoclonal antibodies against influenza B virus
- Author
-
Tadahiro Sasaki, Mutsumi Ito, Ryuta Uraki, Priyanka Soni, Kazuyoshi Ikuta, Yoshihiro Kawaoka, Kiyoko Iwatsuki-Horimoto, Toru Takenaga, Seiya Yamayoshi, and Atsuhiro Yasuhara
- Subjects
0301 basic medicine ,medicine.drug_class ,Hemagglutinin (influenza) ,Hemagglutinin Glycoproteins, Influenza Virus ,Biology ,Monoclonal antibody ,Antibodies, Viral ,Peripheral blood mononuclear cell ,Epitope ,Virus ,hybridoma ,Cell Line ,03 medical and health sciences ,Mice ,Immune system ,Virology ,SPYMEG ,medicine ,Animals ,Humans ,Hybridomas ,General Veterinary ,Full Paper ,Antibodies, Monoclonal ,influenza B virus ,030104 developmental biology ,Cell culture ,biology.protein ,Leukocytes, Mononuclear ,Hybridoma technology ,human monoclonal antibody - Abstract
Influenza B virus has been known to infect humans and other animals, including seals. Vaccination efficacy varies across seasons. Human monoclonal antibodies (mAbs) can be useful for developing novel vaccines, guided by epitope analysis, and can be used therapeutically. Hybridoma technology has been used to make mAbs. Here we evaluated SPYMEG as a fusion partner cell line for human mAb generation specific to influenza B hemagglutinin (HA). SPYMEG is a human/murine myeloma partner cell line that has previously been used to generate human mAbs that recognize the HA of influenza A and B viruses. Peripheral blood mononuclear cells were obtained from 16 volunteers, previously vaccinated with the 2014-2015 trivalent seasonal influenza vaccine, and were fused with SPYMEG to yield hybridomas. The resulting hybridomas were screened for antigen-specific antibody secretion and cloned by limiting dilution. We obtained 32 stable clones secreting anti-influenza B HA human IgG, although most of these clones were obtained from one volunteer (SeaV-29) who had a robust immune response. We conclude that SPYMEG is a good fusion partner cell line, although cloning by limiting dilution may lead to significant loss of hybridomas.
- Published
- 2018
47. Development of an Influenza Rapid Diagnostic Kit Specific for the H7 Subtype
- Author
-
Kiyoko Iwatsuki-Horimoto, Jianzhong Shi, Xiurong Wang, Yuko Sakai-Tagawa, Mutsumi Ito, Kazushi Murakami, Tiago J. da Silva Lopes, Kazunari Nakaishi, Seiya Yamayoshi, Satoshi Watabe, Hualan Chen, and Yoshihiro Kawaoka
- Subjects
0301 basic medicine ,Microbiology (medical) ,medicine.drug_class ,Highly pathogenic ,viruses ,lcsh:QR1-502 ,Early detection ,Biology ,Monoclonal antibody ,medicine.disease_cause ,Microbiology ,Virus ,lcsh:Microbiology ,influenza virus ,Seasonal influenza ,03 medical and health sciences ,Pandemic ,medicine ,highly pathogenic avian influenza ,Original Research ,Virology ,Low pathogenic ,Influenza A virus subtype H5N1 ,030104 developmental biology ,monoclonal antibody ,rapid diagnostic kit ,H7 subtype - Abstract
Since the spring of 2013, human infections with H7N9 viruses have been detected in China. Some of these viruses have become highly pathogenic. Highly and low pathogenic avian influenza H7N9 viruses are currently co-circulating with the seasonal influenza A viruses H3N2 and H1N1pdm09. Prompt identification and isolation of H7N9 patients is one measure to prevent the spread of H7N9 virus and help prevent a pandemic. The majority of commercially available point-of-care rapid influenza diagnostic kits can differentiate between influenza A and B viruses, but cannot distinguish between H7N9 viruses and seasonal influenza A viruses. Accordingly, we have developed a rapid diagnostic kit specific for the H7 subtype that is accessible, easy to use. Although the detection limit of this H7 kit is one-tenth lower than that of a commercially available rapid influenza A and B diagnostic kit of similar design, except for the specificity of the monoclonal antibodies used, this kit is highly specific, detecting only H7-subtype influenza viruses, including the recent highly pathogenic H7N9 viruses from humans, and does not show any non-specific reactions with other HA subtypes. This H7 kit will be of value for the early detection of H7N9-infected patients.
- Published
- 2018
48. Host protein mimics viral protein to hinder infection by Ebola virus
- Author
-
Yoshihiro Kawaoka and Seiya Yamayoshi
- Subjects
0303 health sciences ,Multidisciplinary ,Ebola virus ,Viral protein ,viruses ,virus diseases ,RNA ,Disease ,Biology ,medicine.disease_cause ,Virology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Viral rna ,030217 neurology & neurosurgery ,Host protein ,030304 developmental biology - Abstract
Infection by Ebola virus can be fatal. The discovery of a human protein that mimics one type of Ebola protein and binds to another to suppress viral RNA production might aid the development of clinical treatments for the disease. The production of RNA by Ebola virus is suppressed by a human protein.
- Published
- 2019
49. Diversity of antigenic mutants of influenza A(H1N1)pdm09 virus escaped from human monoclonal antibodies
- Author
-
Kazuyoshi Ikuta, Ryuta Uraki, Shinya Yamada, Yoshihiro Kawaoka, Mutsumi Ito, Tadahiro Sasaki, Emi Takashita, Yuko Sakai-Tagawa, Priyanka Soni, Atsuhiro Yasuhara, Kiyoko Iwatsuki-Horimoto, Seiya Yamayoshi, Toru Takenaga, and Chiharu Kawakami
- Subjects
0301 basic medicine ,medicine.drug_class ,viruses ,Glutamine ,030106 microbiology ,Mutant ,lcsh:Medicine ,Hemagglutinin Glycoproteins, Influenza Virus ,Monoclonal antibody ,medicine.disease_cause ,Antigenic drift ,Virus ,Article ,03 medical and health sciences ,Influenza A Virus, H1N1 Subtype ,Antigen ,Influenza, Human ,medicine ,Influenza A virus ,Humans ,Amino Acid Sequence ,lcsh:Science ,Epidemics ,Antigens, Viral ,Mutation ,Multidisciplinary ,biology ,Lysine ,lcsh:R ,Antibodies, Monoclonal ,Genetic Variation ,Virology ,Antigenic Variation ,030104 developmental biology ,Amino Acid Substitution ,biology.protein ,lcsh:Q ,Antibody - Abstract
Since the 2017 Southern Hemisphere influenza season, the A(H1N1)pdm09-like virus recommended for use in the vaccine was changed because human, but not ferret, sera distinguish A(H1N1)pdm09 viruses isolated after 2013 from the previously circulating strains. An amino acid substitution, lysine to glutamine, at position 166 (H3 numbering) in the major antigenic site of HA was reported to be responsible for the antigenic drift. Here, we obtained two anti-A(H1N1)pdm09 HA monoclonal antibodies that failed to neutralize viruses isolated after 2013 from a vaccinated volunteer. Escape mutations were identified at position 129, 165, or 166 in the major antigenic site of HA. Competitive growth of the escape mutant viruses with the wild-type virus revealed that some escape mutants possessing an amino acid substitution other than K166Q showed superior growth to that of the wild-type virus. These results suggest that in addition to the K166Q mutation that occurred in epidemic strains, other HA mutations can confer resistance to antibodies that recognize the K166 area, leading to emergence of epidemic strains with such mutations.
- Published
- 2017
50. Mapping of a Region of the PA-X Protein of Influenza A Virus That Is Important for Its Shutoff Activity
- Author
-
Kohei Oishi, Seiya Yamayoshi, and Yoshihiro Kawaoka
- Subjects
Molecular Sequence Data ,Immunology ,Viral Nonstructural Proteins ,medicine.disease_cause ,Microbiology ,Open Reading Frames ,Endonuclease ,Virology ,Influenza A virus ,medicine ,Amino Acid Sequence ,Luciferases ,Gene ,Basic amino acids ,chemistry.chemical_classification ,Messenger RNA ,Base Sequence ,biology ,Host (biology) ,Sequence Analysis, DNA ,Endonucleases ,Molecular biology ,Protein Structure, Tertiary ,Genome Replication and Regulation of Viral Gene Expression ,Amino acid ,Repressor Proteins ,chemistry ,Mutagenesis ,Insect Science ,biology.protein ,Function (biology) - Abstract
Influenza A virus PA-X comprises an N-terminal PA endonuclease domain and a C-terminal PA-X-specific domain. PA-X reduces host and viral mRNA accumulation via its endonuclease function. Here, we found that the N-terminal 15 amino acids, particularly six basic amino acids, in the C-terminal PA-X-specific region are important for PA-X shutoff activity. These six basic amino acids enabled a PA deletion mutant to suppress protein expression at a level comparable to that of wild-type PA-X.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.