Network


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

Hotspot


Dive into the research topics where Ryuta Uraki is active.

Publication


Featured researches published by Ryuta Uraki.


Nature | 2013

Characterization of H7N9 influenza A viruses isolated from humans.

Tokiko Watanabe; Maki Kiso; Satoshi Fukuyama; Noriko Nakajima; Masaki Imai; S. Yamada; Shin Murakami; Seiya Yamayoshi; Kiyoko Iwatsuki-Horimoto; Yoshihiro Sakoda; Emi Takashita; Ryan McBride; Takeshi Noda; Masato Hatta; Hirotaka Imai; Dongming Zhao; Noriko Kishida; Masayuki Shirakura; Robert P. de Vries; Shintaro Shichinohe; Masatoshi Okamatsu; Tomokazu Tamura; Yuriko Tomita; Naomi Fujimoto; Kazue Goto; Hiroaki Katsura; Eiryo Kawakami; Izumi Ishikawa; Shinji Watanabe; Mutsumi Ito

Avian influenza A viruses rarely infect humans; however, when human infection and subsequent human-to-human transmission occurs, worldwide outbreaks (pandemics) can result. The recent sporadic infections of humans in China with a previously unrecognized avian influenza A virus of the H7N9 subtype (A(H7N9)) have caused concern owing to the appreciable case fatality rate associated with these infections (more than 25%), potential instances of human-to-human transmission, and the lack of pre-existing immunity among humans to viruses of this subtype. Here we characterize two early human A(H7N9) isolates, A/Anhui/1/2013 (H7N9) and A/Shanghai/1/2013 (H7N9); hereafter referred to as Anhui/1 and Shanghai/1, respectively. In mice, Anhui/1 and Shanghai/1 were more pathogenic than a control avian H7N9 virus (A/duck/Gunma/466/2011 (H7N9); Dk/GM466) and a representative pandemic 2009 H1N1 virus (A/California/4/2009 (H1N1pdm09); CA04). Anhui/1, Shanghai/1 and Dk/GM466 replicated well in the nasal turbinates of ferrets. In nonhuman primates, Anhui/1 and Dk/GM466 replicated efficiently in the upper and lower respiratory tracts, whereas the replicative ability of conventional human influenza viruses is typically restricted to the upper respiratory tract of infected primates. By contrast, Anhui/1 did not replicate well in miniature pigs after intranasal inoculation. Critically, Anhui/1 transmitted through respiratory droplets in one of three pairs of ferrets. Glycan arrays showed that Anhui/1, Shanghai/1 and A/Hangzhou/1/2013 (H7N9) (a third human A(H7N9) virus tested in this assay) bind to human virus-type receptors, a property that may be critical for virus transmissibility in ferrets. Anhui/1 was found to be less sensitive in mice to neuraminidase inhibitors than a pandemic H1N1 2009 virus, although both viruses were equally susceptible to an experimental antiviral polymerase inhibitor. The robust replicative ability in mice, ferrets and nonhuman primates and the limited transmissibility in ferrets of Anhui/1 suggest that A(H7N9) viruses have pandemic potential.


Journal of Virology | 2014

Virulence-Affecting Amino Acid Changes in the PA Protein of H7N9 Influenza A Viruses

Seiya Yamayoshi; S. Yamada; Satoshi Fukuyama; Shin Murakami; Dongming Zhao; Ryuta Uraki; Tokiko Watanabe; Yuriko Tomita; Catherine A. Macken; Gabriele Neumann; Yoshihiro Kawaoka

ABSTRACT Novel avian-origin influenza A(H7N9) viruses were first reported to infect humans in March 2013. To date, 143 human cases, including 45 deaths, have been recorded. By using sequence comparisons and phylogenetic and ancestral inference analyses, we identified several distinct amino acids in the A(H7N9) polymerase PA protein, some of which may be mammalian adapting. Mutant viruses possessing some of these amino acid changes, singly or in combination, were assessed for their polymerase activities and growth kinetics in mammalian and avian cells and for their virulence in mice. We identified several mutants that were slightly more virulent in mice than the wild-type A(H7N9) virus, A/Anhui/1/2013. These mutants also exhibited increased polymerase activity in human cells but not in avian cells. Our findings indicate that the PA protein of A(H7N9) viruses has several amino acid substitutions that are attenuating in mammals. IMPORTANCE Novel avian-origin influenza A(H7N9) viruses emerged in the spring of 2013. By using computational analyses of A(H7N9) viral sequences, we identified several amino acid changes in the polymerase PA protein, which we then assessed for their effects on viral replication in cultured cells and mice. We found that the PA proteins of A(H7N9) viruses possess several amino acid substitutions that cause attenuation in mammals.


Science Advances | 2017

Zika virus causes testicular atrophy

Ryuta Uraki; Jesse Hwang; Kellie Ann Jurado; Sarah Householder; Laura J. Yockey; Andrew K. Hastings; Robert J. Homer; Akiko Iwasaki; Erol Fikrig

Zika virus replicates in mouse testes and causes testicular atrophy, with implication on sexual transmission and male fertility. Zika virus (ZIKV) is an emerging mosquito-borne flavivirus that has recently been found to cause fetal infection and neonatal abnormalities, including microcephaly and neurological dysfunction. ZIKV persists in the semen months after the acute viremic phase in humans. To further understand the consequences of ZIKV persistence in males, we infected Ifnar1−/− mice via subcutaneous injection of a pathogenic but nonlethal ZIKV strain. ZIKV replication persists within the testes even after clearance from the blood, with interstitial, testosterone-producing Leydig cells supporting virus replication. We found high levels of viral RNA and antigen within the epididymal lumen, where sperm is stored, and within surrounding epithelial cells. Unexpectedly, at 21 days post-infection, the testes of the ZIKV-infected mice were significantly smaller compared to those of mock-infected mice, indicating progressive testicular atrophy. ZIKV infection caused a reduction in serum testosterone, suggesting that male fertility can be affected. Our findings have important implications for nonvector-borne vertical transmission, as well as long-term potential reproductive deficiencies, in ZIKV-infected males.


Neuroscience Letters | 2010

Oxidative damage to neurons caused by the induction of microglial NADPH oxidase in encephalomyocarditis virus infection.

Yasuhisa Ano; Tetsuya Kimata; Ryuta Uraki; Katsuaki Sugiura; Takashi Onodera

Reactive oxygen species (ROS) play an important role in diverse vital functions including host defense via anti-viral and anti-bacterial effects, but ROS also lead to peroxynitrite and hydroxyl radical production, which are powerful mediators of brain injury in brain inflammation. It is known that NADPH oxidases (NOX) are the major source of ROS. In the present study, NOX2 expression and distribution were examined after intracranial encephalomyocarditis virus B variant (EMCV-B) infection, which causes encephalitis. The reverse transcriptase (RT)-polymerase chain reaction (PCR) and immunohistochemistry showed that the expression and distribution of NOX2 were significantly up-regulated after EMCV-B infection in microglial cells, which invaded into the surrounding regions where neurons were subjected to oxidative stress. These findings suggest that the oxidative stress generated by NOX2 in activated microglial cells damages neurons and that this is an important process in the pathogenesis of EMCV-B infection.


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.


Scientific Reports | 2013

Protective efficacy of orally administered, heat-killed Lactobacillus pentosus b240 against influenza A virus

Maki Kiso; Ryo Takano; Saori Sakabe; Hiroaki Katsura; Kyoko Shinya; Ryuta Uraki; Shinji Watanabe; Hiroshi Saito; Masamichi Toba; Noriyuki Kohda; Yoshihiro Kawaoka

Influenza A(H1N1)pdm virus caused the first human pandemic of the 21st century. Although various probiotic Lactobacillus species have been shown to have anti-microbial effects against pneumonia-inducing pathogens, the prophylactic efficacy and mechanisms behind their protection remain largely unknown. Here, we evaluated the prophylactic efficacy of heat-killed Lactobacillus pentosus b240 against lethal influenza A(H1N1)pdm virus infection in a mouse model. To further define the protective responses induced by b240, we performed virologic, histopathologic, and transcriptomic analyses on the mouse lungs. Although we did not observe an appreciable effect of b240 on virus growth, cytokine production, or histopathology, gene expressional analysis revealed that oral administration of b240 differentially regulates antiviral gene expression in mouse lungs. Our results unveil the possible mechanisms behind the protection mediated by b240 against influenza virus infection and provide new insights into probiotic therapy.


Journal of Virology | 2013

A Novel Bivalent Vaccine Based on a PB2-Knockout Influenza Virus Protects Mice from Pandemic H1N1 and Highly Pathogenic H5N1 Virus Challenges

Ryuta Uraki; Maki Kiso; Kiyoko Iwatsuki-Horimoto; Satoshi Fukuyama; Emi Takashita; Makoto Ozawa; Yoshihiro Kawaoka

ABSTRACT Vaccination is an effective means to protect against influenza virus. Although inactivated and live-attenuated vaccines are currently available, each vaccine has disadvantages (e.g., immunogenicity and safety issues). To overcome these problems, we previously developed a replication-incompetent PB2-knockout (PB2-KO) influenza virus that replicates only in PB2 protein-expressing cells. Here, we generated two PB2-KO viruses whose PB2-coding regions were replaced with the HA genes of either A/California/04/2009 (H1N1pdm09) or A/Vietnam/1203/2004 (H5N1). The resultant viruses comparably, or in some cases more efficiently, induced virus-specific antibodies in the serum, nasal wash, and bronchoalveolar lavage fluid of mice relative to a conventional formalin-inactivated vaccine. Furthermore, mice immunized with these PB2-KO viruses were protected from lethal challenges with not only the backbone virus strain but also strains from which their foreign HAs originated, indicating that PB2-KO viruses with antigenically different HAs could serve as bivalent influenza vaccines.


Journal of Virology | 2013

Virulence Determinants of Pandemic A(H1N1)2009 Influenza Virus in a Mouse Model

Ryuta Uraki; Maki Kiso; Kyoko Shinya; Hideo Goto; Ryo Takano; Kiyoko Iwatsuki-Horimoto; Kazuo Takahashi; Rod S. Daniels; Olav Hungnes; Tokiko Watanabe; Yoshihiro Kawaoka

ABSTRACT A novel swine-origin H1N1 influenza virus [A(H1N1)pdm09 virus] caused the 2009 influenza pandemic. Most patients exhibited mild symptoms similar to seasonal influenza, but some experienced severe clinical signs and, in the worst cases, died. Such differences in symptoms are generally associated with preexisting medical conditions, but recent reports indicate the possible involvement of viral factors in clinical severity. To better understand the mechanism of pathogenicity of the A(H1N1)pdm09 virus, here, we compared five viruses that are genetically similar but were isolated from patients with either severe or mild symptoms. In a mouse model, A/Norway/3487/2009 (Norway3487) virus exhibited greater pathogenicity than did A/Osaka/164/2009 (Osaka164) virus. By exploiting reassortant viruses between these two viruses, we found that viruses possessing the hemagglutinin (HA) gene of Norway3487 in the genetic background of Osaka164 were more pathogenic in mice than other reassortant viruses, indicating a role for HA in the high virulence of Norway3487 virus. Intriguingly, a virus possessing HA, NA, and NS derived from Norway3487 exhibited greater pathogenicity in mice in concert with PB2 and PB1 derived from Osaka164 than did the parental Norway3487 virus. These findings demonstrate that reassortment between A(H1N1)pdm09 viruses can lead to increased pathogenicity and highlight the need for continued surveillance of A(H1N1)pdm09 viruses.


Scientific Reports | 2015

Amino acids substitutions in the PB2 protein of H7N9 influenza A viruses are important for virulence in mammalian hosts

Seiya Yamayoshi; Satoshi Fukuyama; S. Yamada; Dongming Zhao; Shin Murakami; Ryuta Uraki; Tokiko Watanabe; Yuriko Tomita; Gabriele Neumann; Yoshihiro Kawaoka

We tested the biological significance of two amino acid mutations in the PB2 protein (glutamic acid to lysine at position 627 and aspartic acid to asparagine at position 701) of A(H7N9) viruses for mammalian adaptation. Mutants were assessed for their viral polymerase activities, growth kinetics in mammalian and avian cells, and pathogenicity in mice. We found that lysine at position 627 and asparagine at position 701 in PB2 are essential for mammalian adaptation of A(H7N9) viruses.


The Journal of Infectious Diseases | 2017

Fetal Growth Restriction Caused by Sexual Transmission of Zika Virus in Mice

Ryuta Uraki; Kellie Ann Jurado; Jesse Hwang; Klara Szigeti-Buck; Tamas L. Horvath; Akiko Iwasaki; Erol Fikrig

Zika virus (ZIKV) can be transmitted by mosquito bite or sexual contact. Using mice that lack the type I interferon receptor, we examined sexual transmission of ZIKV. Electron microscopy analyses showed association of virions with developing sperm within testes as well as with mature sperm within epididymis. When ZIKV-infected male mice were mated with naive female mice, the weight of fetuses at embryonic day 18.5 was significantly reduced compared with the control group. Additionally, we found ocular deformities in a minority of the fetuses. These results suggest that ZIKV causes fetal abnormalities after female mating with an infected male.

Collaboration


Dive into the Ryuta Uraki's collaboration.

Top Co-Authors

Avatar

Yoshihiro Kawaoka

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge