Network


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

Hotspot


Dive into the research topics where Hirohito Ishigaki is active.

Publication


Featured researches published by Hirohito Ishigaki.


Nature | 2009

In vitro and in vivo characterization of new swine-origin H1N1 influenza viruses

Yasushi Itoh; Kyoko Shinya; Maki Kiso; Tokiko Watanabe; Yoshihiro Sakoda; Masato Hatta; Yukiko Muramoto; Daisuke Tamura; Yuko Sakai-Tagawa; Takeshi Noda; Saori Sakabe; Masaki Imai; Yasuko Hatta; Shinji Watanabe; Chengjun Li; S. Yamada; Ken Fujii; Shin Murakami; Hirotaka Imai; Satoshi Kakugawa; Mutsumi Ito; Ryo Takano; Kiyoko Iwatsuki-Horimoto; Masayuki Shimojima; Taisuke Horimoto; Hideo Goto; Kei Takahashi; Akiko Makino; Hirohito Ishigaki; Misako Nakayama

Influenza A viruses cause recurrent outbreaks at local or global scale with potentially severe consequences for human health and the global economy. Recently, a new strain of influenza A virus was detected that causes disease in and transmits among humans, probably owing to little or no pre-existing immunity to the new strain. On 11 June 2009 the World Health Organization declared that the infections caused by the new strain had reached pandemic proportion. Characterized as an influenza A virus of the H1N1 subtype, the genomic segments of the new strain were most closely related to swine viruses. Most human infections with swine-origin H1N1 influenza viruses (S-OIVs) seem to be mild; however, a substantial number of hospitalized individuals do not have underlying health issues, attesting to the pathogenic potential of S-OIVs. To achieve a better assessment of the risk posed by the new virus, we characterized one of the first US S-OIV isolates, A/California/04/09 (H1N1; hereafter referred to as CA04), as well as several other S-OIV isolates, in vitro and in vivo. In mice and ferrets, CA04 and other S-OIV isolates tested replicate more efficiently than a currently circulating human H1N1 virus. In addition, CA04 replicates efficiently in non-human primates, causes more severe pathological lesions in the lungs of infected mice, ferrets and non-human primates than a currently circulating human H1N1 virus, and transmits among ferrets. In specific-pathogen-free miniature pigs, CA04 replicates without clinical symptoms. The assessment of human sera from different age groups suggests that infection with human H1N1 viruses antigenically closely related to viruses circulating in 1918 confers neutralizing antibody activity to CA04. Finally, we show that CA04 is sensitive to approved and experimental antiviral drugs, suggesting that these compounds could function as a first line of defence against the recently declared S-OIV pandemic.


Journal of Virology | 2014

Disease Severity Is Associated with Differential Gene Expression at the Early and Late Phases of Infection in Nonhuman Primates Infected with Different H5N1 Highly Pathogenic Avian Influenza Viruses

Yukiko Muramoto; Jason E. Shoemaker; Mai Quynh Le; Yasushi Itoh; Daisuke Tamura; Yuko Sakai-Tagawa; Hirotaka Imai; Ryuta Uraki; Ryo Takano; Eiryo Kawakami; Mutsumi Ito; Kiyoko Okamoto; Hirohito Ishigaki; Hitomi Mimuro; Chihiro Sasakawa; Yukiko Matsuoka; Takeshi Noda; Satoshi Fukuyama; Kazumasa Ogasawara; Hiroaki Kitano; Yoshihiro Kawaoka

ABSTRACT Occasional transmission of highly pathogenic avian H5N1 influenza viruses to humans causes severe pneumonia with high mortality. To better understand the mechanisms via which H5N1 viruses induce severe disease in humans, we infected cynomolgus macaques with six different H5N1 strains isolated from human patients and compared their pathogenicity and the global host responses to the virus infection. Although all H5N1 viruses replicated in the respiratory tract, there was substantial heterogeneity in their replicative ability and in the disease severity induced, which ranged from asymptomatic to fatal. A comparison of global gene expression between severe and mild disease cases indicated that interferon-induced upregulation of genes related to innate immunity, apoptosis, and antigen processing/presentation in the early phase of infection was limited in severe disease cases, although interferon expression was upregulated in both severe and mild cases. Furthermore, coexpression analysis of microarray data, which reveals the dynamics of host responses during the infection, demonstrated that the limited expression of these genes early in infection led to a failure to suppress virus replication and to the hyperinduction of genes related to immunity, inflammation, coagulation, and homeostasis in the late phase of infection, resulting in a more severe disease. Our data suggest that the attenuated interferon-induced activation of innate immunity, apoptosis, and antigen presentation in the early phase of H5N1 virus infection leads to subsequent severe disease outcome. IMPORTANCE Highly pathogenic avian H5N1 influenza viruses sometimes transmit to humans and cause severe pneumonia with ca. 60% lethality. The continued circulation of these viruses poses a pandemic threat; however, their pathogenesis in mammals is not fully understood. We, therefore, investigated the pathogenicity of six H5N1 viruses and compared the host responses of cynomolgus macaques to the virus infection. We identified differences in the viral replicative ability of and in disease severity caused by these H5N1 viruses. A comparison of global host responses between severe and mild disease cases identified the limited upregulation of interferon-stimulated genes early in infection in severe cases. The dynamics of the host responses indicated that the limited response early in infection failed to suppress virus replication and led to hyperinduction of pathological condition-related genes late in infection. These findings provide insight into the pathogenesis of H5N1 viruses in mammals.


Antimicrobial Agents and Chemotherapy | 2011

Efficacy of Single Intravenous Injection of Peramivir against Influenza B Virus Infection in Ferrets and Cynomolgus Macaques

Mitsutaka Kitano; Yasushi Itoh; Makoto Kodama; Hirohito Ishigaki; Misako Nakayama; Hideaki Ishida; Kaoru Baba; Takahiro Noda; Kenji Sato; Yoichiro Nihashi; Takushi Kanazu; Ryu Yoshida; Ryuzo Torii; Akihiko Sato; Kazumasa Ogasawara

ABSTRACT We evaluated the efficacy of a single intravenous dose peramivir for treatment of influenza B virus infection in ferrets and cynomolgus macaques in the present study. A single dose of peramivir (60 mg/kg of body weight) given to ferrets on 1 day postinfection with influenza B virus significantly reduced median area under the curve (AUC) virus titers (peramivir, 8.3 log10 50% tissue culture infective doses [TCID50s]·day/ml; control, 10.7 log10 TCID50s·day/ml; P < 0.0001). Furthermore, nasal virus titers on day 2 postinfection in ferrets receiving a single injection of peramivir (30 mg/kg) and AUCs of the body temperature increase in ferrets receiving a single injection of peramivir (30 and 60 mg/kg) were lower than those in ferrets administered oral oseltamivir phosphate (30 and 60 mg/kg/day twice daily for 3 days). In macaques infected with influenza B virus, viral titers in the nasal swab fluid on days 2 and 3 postinfection and body temperature after a single injection of peramivir (30 mg/kg) were lower than those after oral administration of oseltamivir phosphate (30 mg/kg/day for 5 days). The two animal models used in the present study demonstrated that inhibition of viral replication at the early time point after infection was critical in reduction of AUCs of virus titers and interleukin-6 production, resulting in amelioration of symptoms. Our results shown in animal models suggest that the early treatment with a single intravenous injection of peramivir is clinically recommended to reduce symptoms effectively in influenza B virus infection.


Antimicrobial Agents and Chemotherapy | 2015

Emergence of H7N9 Influenza A Virus Resistant to Neuraminidase Inhibitors in Nonhuman Primates

Yasushi Itoh; Shintaro Shichinohe; Misako Nakayama; Manabu Igarashi; Akihiro Ishii; Hirohito Ishigaki; Hideaki Ishida; Naoko Kitagawa; Takako Sasamura; Masanori Shiohara; Michiko Doi; Hideaki Tsuchiya; Shinichiro Nakamura; Masatoshi Okamatsu; Yoshihiro Sakoda; Hiroshi Kida; Kazumasa Ogasawara

ABSTRACT The number of patients infected with H7N9 influenza virus has been increasing since 2013. We examined the efficacy of neuraminidase (NA) inhibitors and the efficacy of a vaccine against an H7N9 influenza virus, A/Anhui/1/2013 (H7N9), isolated from a patient in a cynomolgus macaque model. NA inhibitors (oseltamivir and peramivir) barely reduced the total virus amount because of the emergence of resistant variants with R289K or I219T in NA [residues 289 and 219 in N9 of A/Anhui/1/2013 (H7N9) correspond to 292 and 222 in N2, respectively] in three of the six treated macaques, whereas subcutaneous immunization of an inactivated vaccine derived from A/duck/Mongolia/119/2008 (H7N9) prevented propagation of A/Anhui/1/2013 (H7N9) in all vaccinated macaques. The percentage of macaques in which variant H7N9 viruses with low sensitivity to the NA inhibitors were detected was much higher than that of macaques in which variant H5N1 highly pathogenic influenza virus was detected after treatment with one of the NA inhibitors in our previous study. The virus with R289K in NA was reported in samples from human patients, whereas that with I219T in NA was identified for the first time in this study using macaques, though no variant H7N9 virus was reported in previous studies using mice. Therefore, the macaque model enables prediction of the frequency of emerging H7N9 virus resistant to NA inhibitors in vivo. Since H7N9 strains resistant to NA inhibitors might easily emerge compared to other influenza viruses, monitoring of the emergence of variants is required during treatment of H7N9 influenza virus infection with NA inhibitors.


Vaccine | 2010

Subcutaneous inoculation of a whole virus particle vaccine prepared from a non-pathogenic virus library induces protective immunity against H7N7 highly pathogenic avian influenza virus in cynomolgus macaques

Yasushi Itoh; Hiroichi Ozaki; Hirohito Ishigaki; Yoshihiro Sakoda; Tomoya Nagata; Kosuke Soda; Norikazu Isoda; Taichiro Miyake; Hideaki Ishida; Kiyoko Okamoto; Misako Nakayama; Hideaki Tsuchiya; Ryuzo Torii; Hiroshi Kida; Kazumasa Ogasawara

Development of H7N7 highly pathogenic avian influenza virus (HPAIV) vaccines is an urgent issue since human cases of infection with this subtype virus have been reported and most humans have no immunity against H7N7 viruses. We made an H7N7 vaccine combining components from an influenza virus library of non-pathogenic type A influenza viruses. Antibody and T cell recall responses specific against the vaccine strain were elicited by subcutaneous inoculation with the whole virus particle vaccine with or without alum as an adjuvant in cynomolgus macaques. No significant difference was observed in magnitude of antibody responses between vaccination with alum and vaccination without alum, though vaccination with alum induced longer recall responses of CD8(+) T cells than did vaccination without alum. After challenge with a subtype of H7N7 HPAIV, the virus was detected in nasal swabs of unvaccinated macaques for 8 days but only for 1 day in the animals vaccinated either with or without alum, although the macaques vaccinated with alum showed elevated body temperature more briefly after infection. These findings demonstrated that this H7N7 HPAIV strain is pathogenic to macaques and that the vaccine conferred protective immunity to macaques against H7N7 HPAIV infection.


Microbiology and Immunology | 2008

Analysis of splenic Gr-1int immature myeloid cells in tumor-bearing mice

Yoshiko Yamamoto; Hirohito Ishigaki; Hideaki Ishida; Yasushi Itoh; Yoichi Noda; Kazumasa Ogasawara

It is known that the number of ImC, expressing myeloid markers, CD11b and Gr‐1, increase with tumor growth and ImC play a role in the escape of tumor cells from immunosurveillance in tumor‐bearing mice and cancer patients. However, the mechanisms by which ImC suppress immune responses in tumor‐bearing mice have not been completely elucidated. In the present study, we investigated the function of splenic ImC freshly isolated from tumor‐bearing mice and splenic ImC differentiated in vitro by GM‐CSF. Freshly isolated splenic ImC were divided into two groups depending on Gr‐1 expression, Gr‐1 high (Gr‐1hi) and intermediate (Gr‐1int). Freshly isolated splenic Gr‐1int ImC, but not Gr‐1hi ImC, from tumor‐bearing mice reduced production of IFN‐γ in CD8+ T cells, but neither splenic Gr‐1int ImC nor Gr‐1hi ImC isolated from naive mice did. Both Gr‐1int and Gr‐1hi ImC differentiated in vitro by GM‐CSF inhibited production of IFN‐γ in both CD8+ and CD4+ T cells. In addition, the differentiated Gr‐1int ImC, one‐third of which were CD11c+F4/80+ cells, and their culture supernatants suppressed proliferative responses of T cells stimulated by CD3 ligation, but the differentiated Gr‐1hi ImC and their culture supernatants did not. These results suggest that Gr‐1int ImC are altered to immune‐suppressive cells in tumor circumstances and that they are differentiated by GM‐CSF progressively into CD11c+F4/80+ cells with further suppressive activity against T cells.


PLOS Pathogens | 2014

Protective Efficacy of Passive Immunization with Monoclonal Antibodies in Animal Models of H5N1 Highly Pathogenic Avian Influenza Virus Infection

Yasushi Itoh; Reiko Yoshida; Shintaro Shichinohe; Megumi Higuchi; Hirohito Ishigaki; Misako Nakayama; Van Loi Pham; Hideaki Ishida; Mitsutaka Kitano; Masahiko Arikata; Naoko Kitagawa; Yachiyo Mitsuishi; Kazumasa Ogasawara; Hideaki Tsuchiya; Takahiro Hiono; Masatoshi Okamatsu; Yoshihiro Sakoda; Hiroshi Kida; Mutsumi Ito; Le Quynh Mai; Yoshihiro Kawaoka; Hiroko Miyamoto; Mari Ishijima; Manabu Igarashi; Yasuhiko Suzuki; Ayato Takada

Highly pathogenic avian influenza (HPAI) viruses of the H5N1 subtype often cause severe pneumonia and multiple organ failure in humans, with reported case fatality rates of more than 60%. To develop a clinical antibody therapy, we generated a human-mouse chimeric monoclonal antibody (MAb) ch61 that showed strong neutralizing activity against H5N1 HPAI viruses isolated from humans and evaluated its protective potential in mouse and nonhuman primate models of H5N1 HPAI virus infections. Passive immunization with MAb ch61 one day before or after challenge with a lethal dose of the virus completely protected mice, and partial protection was achieved when mice were treated 3 days after the challenge. In a cynomolgus macaque model, reduced viral loads and partial protection against lethal infection were observed in macaques treated with MAb ch61 intravenously one and three days after challenge. Protective effects were also noted in macaques under immunosuppression. Though mutant viruses escaping from neutralization by MAb ch61 were recovered from macaques treated with this MAb alone, combined treatment with MAb ch61 and peramivir reduced the emergence of escape mutants. Our results indicate that antibody therapy might be beneficial in reducing viral loads and delaying disease progression during H5N1 HPAI virus infection in clinical cases and combined treatment with other antiviral compounds should improve the protective effects of antibody therapy against H5N1 HPAI virus infection.


Nature Communications | 2017

MHC matching improves engraftment of iPSC-derived neurons in non-human primates

Asuka Morizane; Tetsuhiro Kikuchi; Takuya Hayashi; Hiroshi Mizuma; Sayuki Takara; Hisashi Doi; Aya Mawatari; Matthew F. Glasser; Takashi Shiina; Hirohito Ishigaki; Yasushi Itoh; Keisuke Okita; Emi Yamasaki; Daisuke Doi; Hirotaka Onoe; Kazumasa Ogasawara; Shinya Yamanaka; Jun Takahashi

The banking of human leukocyte antigen (HLA)-homozygous-induced pluripotent stem cells (iPSCs) is considered a future clinical strategy for HLA-matched cell transplantation to reduce immunological graft rejection. Here we show the efficacy of major histocompatibility complex (MHC)-matched allogeneic neural cell grafting in the brain, which is considered a less immune-responsive tissue, using iPSCs derived from an MHC homozygous cynomolgus macaque. Positron emission tomography imaging reveals neuroinflammation associated with an immune response against MHC-mismatched grafted cells. Immunohistological analyses reveal that MHC-matching reduces the immune response by suppressing the accumulation of microglia (Iba-1+) and lymphocytes (CD45+) into the grafts. Consequently, MHC-matching increases the survival of grafted dopamine neurons (tyrosine hydroxylase: TH+). The effect of an immunosuppressant, Tacrolimus, is also confirmed in the same experimental setting. Our results demonstrate the rationale for MHC-matching in neural cell grafting to the brain and its feasibility in a clinical setting.Major histocompatibility complex (MHC) matching improves graft survival rates after organ transplantation. Here the authors show that in macaques, MHC-matched iPSC-derived neurons provide better engraftment in the brain, with a lower immune response and higher survival of the transplanted neurons.


PLOS ONE | 2013

Pathogenicity of pandemic H1N1 influenza A virus in immunocompromised cynomolgus macaques

Van Loi Pham; Misako Nakayama; Yasushi Itoh; Hirohito Ishigaki; Mitsutaka Kitano; Masahiko Arikata; Hideaki Ishida; Naoko Kitagawa; Shintaro Shichinohe; Masatoshi Okamatsu; Yoshihiro Sakoda; Hideaki Tsuchiya; Shinichiro Nakamura; Hiroshi Kida; Kazumasa Ogasawara

Pandemic (H1N1) 2009 influenza virus spread throughout the world since most people did not have immunity against the virus. In the post pandemic phase when many humans might possess immunity against the pandemic virus, one of the concerns is infection in immunocompromised people. Therefore, we used an immunosuppressed macaque model to examine pathogenicity of the pandemic (H1N1) 2009 virus under an immunocompromised condition. The virus in nasal samples of immunosuppressed macaques infected with the pandemic (H1N1) 2009 virus was detected longer after infection than was the virus in nasal samples of immunocompetent macaques. As expected, not only virus amounts but also virus propagation sites in the immunosuppressed macaques were larger than those in lungs of the immunocompetent macaques when they were infected with the pandemic virus. Immunosuppressed macaques possessed low levels of immune cells producing cytokines and chemokines, but levels of inflammatory cytokines/chemokine interleukin (IL)-6, IL-18, and monocyte chemotactic protein (MCP)-1 in lungs of the immunosuppressed macaques were higher than those in lungs of the immunocompetent macaques, though the differences were not statistically significant. Therefore, under an immunosuppressive condition, the pandemic influenza (H1N1) 2009 virus might cause more severe morbidity with high cytokine/chemokine production by the host innate immune system than that seen in macaques under the immunocompetent condition.


Journal of Medical Primatology | 2010

Amelioration of pneumonia with Streptococcus pneumoniae infection by inoculation with a vaccine against highly pathogenic avian influenza virus in a non-human primate mixed infection model

Taichiro Miyake; Kosuke Soda; Yasushi Itoh; Yoshihiro Sakoda; Hirohito Ishigaki; Tomoya Nagata; Hideaki Ishida; Misako Nakayama; Hiroichi Ozaki; Hideaki Tsuchiya; Ryuzo Torii; Hiroshi Kida; Kazumasa Ogasawara

Background  Highly pathogenic avian influenza virus (HPAIV) infection has a high mortality rate in humans. Secondary bacterial pneumonia with HPAIV infection has not been reported in human patients, whereas seasonal influenza viruses sometimes enhance bacterial pneumonia, resulting in substantial morbidity and mortality. Therefore, if HPAIV infection were accompanied by bacterial infection, an increase in mortality would be expected. We examined whether a vaccine against HPAIV prevents severe morbidity caused by mixed infection with HPAIV and bacteria.

Collaboration


Dive into the Hirohito Ishigaki's collaboration.

Top Co-Authors

Avatar

Kazumasa Ogasawara

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Yasushi Itoh

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Misako Nakayama

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar

Hideaki Ishida

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hideaki Tsuchiya

Shiga University of Medical Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge