Network


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

Hotspot


Dive into the research topics where Hiroaki Yoshii is active.

Publication


Featured researches published by Hiroaki Yoshii.


Immunity | 2014

The Transcription Factor IRF8 Activates Integrin-Mediated TGF-β Signaling and Promotes Neuroinflammation

Yuko Yoshida; Ryusuke Yoshimi; Hiroaki Yoshii; Daniel Kim; Anup Dey; Huabao Xiong; Jeeva Munasinghe; Itaru Yazawa; Michael J. Donovan; Olga A. Maximova; Suveena Sharma; Hongsheng Wang; Herbert C. Morse; Keiko Ozato

Recent epidemiological studies have identified interferon regulatory factor 8 (IRF8) as a susceptibility factor for multiple sclerosis (MS). However, how IRF8 influences the neuroinflammatory disease has remained unknown. By studying the role of IRF8 in experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, we found that Irf8(-/-) mice are resistant to EAE. Furthermore, expression of IRF8 in antigen-presenting cells (APCs, such as macrophages, dendritic cells, and microglia), but not in T cells, facilitated disease onset and progression through multiple pathways. IRF8 enhanced αvβ8 integrin expression in APCs and activated TGF-β signaling leading to T helper 17 (Th17) cell differentiation. IRF8 induced a cytokine milieu that favored growth and maintenance of Th1 and Th17 cells, by stimulating interleukin-12 (IL-12) and IL-23 production, but inhibiting IL-27 during EAE. Finally, IRF8 activated microglia and exacerbated neuroinflammation. Together, this work provides mechanistic bases by which IRF8 contributes to the pathogenesis of MS.


Proceedings of the National Academy of Sciences of the United States of America | 2009

HIV-1 Vif-mediated ubiquitination/degradation of APOBEC3G involves four critical lysine residues in its C-terminal domain

Yasumasa Iwatani; Denise S.B. Chan; Lin Liu; Hiroaki Yoshii; Junko Shibata; Naoki Yamamoto; Judith G. Levin; Angela M. Gronenborn; Wataru Sugiura

During coevolution with the host, HIV-1 developed the ability to hijack the cellular ubiquitin/proteasome degradation pathway to counteract the antiviral activity of APOBEC3G (A3G), a host cytidine deaminase that can block HIV-1 replication. Abrogation of A3G function involves the HIV-1 Vif protein, which binds A3G and serves as an adapter molecule to recruit A3G to a Cullin5-based E3 ubiquitin ligase complex. Structure-guided mutagenesis of A3G focused on the 14 most surface-exposed Lys residues allowed us to identify four Lys residues (Lys-297, 301, 303, and 334) that are required for Vif-mediated A3G ubiquitination and degradation. Substitution of Arg for these residues confers Vif resistance and restores A3Gs antiviral activity in the presence of Vif. In our model, the critical four Lys residues cluster at the C terminus, opposite to the known N-terminal Vif-interaction region in the protein. Thus, spatial constraints imposed by the E3 ligase complex may be an important determinant in Vif-dependent A3G ubiquitination.


Virology | 2008

Ezrin, Radixin, and Moesin (ERM) proteins function as pleiotropic regulators of human immunodeficiency virus type 1 infection.

Yoshinao Kubo; Hiroaki Yoshii; Haruka Kamiyama; Chika Tominaga; Yuetsu Tanaka; Hironori Sato; Naoki Yamamoto

Ezrin, radixin, and moesin (ERM) proteins supply functional linkage between integral membrane proteins and cytoskeleton in mammalian cells to regulate membrane protein dynamisms and cytoskeleton rearrangement. To assess potential role of the ERM proteins in HIV-1 lifecycle, we examined if suppression of ERM function in human cells expressing HIV-1 infection receptors influences HIV-1 envelope (Env)-mediated HIV-1-vector transduction and cell-cell fusion. Expression of an ezrin dominant negative mutant or knockdown of ezrin, radixin, or moesin with siRNA uniformly decreased transduction titers of HIV-1 vectors having X4-tropic Env. In contrast, transduction titers of R5-tropic Env HIV-1 vectors were decreased only by radixin knockdown: ezrin knockdown had no detectable effects and moesin knockdown rather increased transduction titer. Each of the ERM suppressions had no detectable effects on cell surface expression of CD4, CCR5, and CXCR4 or VSV-Env-mediated HIV-1 vector transductions. Finally, the individual knockdown of ERM mRNAs uniformly decreased efficiency of cell-cell fusion mediated by X4- or R5-tropic Env and HIV-1 infection receptors. These results suggest that (i) the ERM proteins function as positive regulators of infection by X4-tropic HIV-1, (ii) moesin additionally functions as a negative regulator of R5-tropic HIV-1 virus infection at the early step(s) after the membrane fusion, and (iii) receptor protein dynamisms are regulated differently in R5- and X4-tropic HIV-1 infections.


Virology | 2009

Raft localization of CXCR4 is primarily required for X4-tropic human immunodeficiency virus type 1 infection.

Haruka Kamiyama; Hiroaki Yoshii; Yuetsu Tanaka; Hironori Sato; Naoki Yamamoto; Yoshinao Kubo

Human immunodeficiency virus type 1 (HIV-1) infection is initiated by successive interactions of viral envelope glycoprotein gp120 with two cellular surface proteins, CD4 and chemokine receptor. The two most common chemokine receptors that allow HIV-1 entry are the CCR5 and CXCR4. The CD4 and CCR5 are mainly localized to the particular plasma membrane microdomains, termed raft, which is rich in glycolipids and cholesterol. However, the CXCR4 is localized only partially to the raft region. Although the raft domain is suggested to participate in HIV-1 infection, its role in entry of CXCR4-tropic (X4-tropic) virus is still unclear. Here, we used a combination of CD4-independent infection system and cholesterol-depletion-inducing reagent, methyl-beta-cyclodextrin (MbetaCD), to address the requirement of raft domain in the X4-tropic virus infection. Treatment of CD4-negative, CXCR4-positive human cells with MbetaCD inhibited CD4-independent infection of the X4-tropic strains. This inhibitory effect of the cholesterol depletion was observed even when the CXCR4 was over-expressed on the target cells. Soluble CD4-induced infection was also inhibited by MbetaCD. The MbetaCD had no effect on the levels of cell surface expression of CXCR4. In contrast to these infections, MbetaCD treatment did not inhibit CD4-dependent HIV-1 infection in the wild type CD4-expressing cells. This study and previous reports showing that CD4 mutants localized to non-raft domains function as HIV-1 receptor indicate that CXCR4 clustering in the raft microdomains, rather than CD4, is the key step for the HIV-1 entry.


PLOS ONE | 2011

CD4-independent human immunodeficiency virus infection involves participation of endocytosis and cathepsin B.

Hiroaki Yoshii; Haruka Kamiyama; Kensuke Goto; Kazunori Oishi; Nobuhiko Katunuma; Yuetsu Tanaka; Hideki Hayashi; Toshifumi Matsuyama; Hironori Sato; Naoki Yamamoto; Yoshinao Kubo

During a comparison of the infectivity of mNDK, a CD4-independent human immunodeficiency virus type 1 (HIV-1) strain, to various cell lines, we found that HeLa cells were much less susceptible than 293T and TE671 cells. Hybridoma cells between HeLa and 293T cells were as susceptible as 293T cells, suggesting that cellular factors enhance the mNDK infection in 293T cells. By screening a cDNA expression library in HeLa cells, cystatin C was isolated as an enhancer of the mNDK infection. Because cathepsin B protease, a natural ligand of cystatin C, was upregulated in HeLa cells, we speculated that the high levels of cathepsin B activities were inhibitory to the CD4-independent infection and that cystatin C enhanced the infection by impairing the excessive cathepsin B activity. Consistent with this idea, pretreatment of HeLa cells with 125 µM of CA-074Me, a cathepsin B inhibitor, resulted in an 8-fold enhancement of the mNDK infectivity. Because cathepsin B is activated by low pH in acidic endosomes, we further examined the potential roles of endosomes in the CD4-independent infection. Suppression of endosome acidification or endocytosis by inhibitors or by an Eps15 dominant negative mutant reduced the infectivity of mNDK in which CD4-dependent infections were not significantly impaired. Taken together, these results suggest that endocytosis, endosomal acidification, and cathepsin B activity are involved in the CD4-independent entry of HIV-1.


Virology | 2009

Cathepsin L is required for ecotropic murine leukemia virus infection in NIH3T3 cells

Hiroaki Yoshii; Haruka Kamiyama; Kazuo Minematsu; Kensuke Goto; Tsutomu Mizota; Kazunori Oishi; Nobuhiko Katunuma; Naoki Yamamoto; Yoshinao Kubo

Abstract Recently it has been reported that a cathepsin B inhibitor, CA-074Me, attenuates ecotropic murine leukemia virus (Eco-MLV) infection in NIH3T3 cells, suggesting that cathepsin B is required for the Eco-MLV infection. However, cathepsin B activity was negative or extremely low in NIH3T3 cells. How did CA-074Me attenuate the Eco-MLV infection? The CA-074Me treatment of NIH3T3 cells inhibited cathepsin L activity, and a cathepsin L specific inhibitor, CLIK148, attenuated the Eco-MLV vector infection. These results indicate that the suppression of cathepsin L activity by CA-074Me induces the inhibition of Eco-MLV infection, suggesting that cathepsin L is required for the Eco-MLV infection in NIH3T3 cells. The CA-074Me treatment inhibited the Eco-MLV infection in human cells expressing the exogenous mouse ecotropic receptor and endogenous cathepsins B and L, but the CLIK148 treatment did not, showing that only the cathepsin L suppression by CLIK148 is not enough to prevent the Eco-MLV infection in cells expressing both of cathepsins B and L, and CA-074Me inhibits the Eco-MLV infection by suppressing both of cathepsins B and L. These results suggest that either cathepsin B or L is sufficient for the Eco-MLV infection.


Archives of Virology | 2007

Characterization of R peptide of murine leukemia virus envelope glycoproteins in syncytium formation and entry

Yoshinao Kubo; Chika Tominaga; Hiroaki Yoshii; Haruka Kamiyama; Chiho Mitani; H. Amanuma; Naoki Yamamoto

SummaryThe C-terminal R peptide of ecotropic murine leukemia virus (MLV) envelope protein (Env) negatively controls membrane fusion activity. The R peptide cleavage during virion maturation activates its fusogenicity and is required for viral entry. We analyzed fusogenicity and transduction efficiency of mutant Env proteins of ecotropic, amphotropic, polytropic, and xenotropic MLVs. As the result, we found that the hydrophobic amino acid residues around the R peptide cleavage site are important for membrane fusion inhibition by the R peptide. In addition, we found that Env complexes with R peptide-truncated and -containing Env proteins have lower fusogenicity and transduction efficiency than those with the R-peptide-truncated Env alone, suggesting that efficient R peptide cleavage is required for efficient MLV vector transduction. The role of R peptide cleavage in amphotropic, polytropic, and xenotropic MLV infection has not been investigated. We found in this study that the R peptide cleavage is required for amphotropic, xenotropic, and polytropic MLV vector transduction, like with ecotropic MLV. The R-peptide-truncated Env proteins of the xenotropic and polytropic MLVs, however, had much lower fusogenicity than those of the ecotropic and amphotropic MLVs. These results provide valuable information for construction of efficient MLV vectors and for understanding the retroviral entry mechanism.


PLOS ONE | 2011

Infection of XC Cells by MLVs and Ebola Virus Is Endosome-Dependent but Acidification-Independent

Haruka Kamiyama; Katsura Kakoki; Hiroaki Yoshii; Masatomo Iwao; Tsukasa Igawa; Hideki Sakai; Hideki Hayashi; Toshifumi Matsuyama; Naoki Yamamoto; Yoshinao Kubo

Inhibitors of endosome acidification or cathepsin proteases attenuated infections mediated by envelope proteins of xenotropic murine leukemia virus-related virus (XMRV) and Ebola virus, as well as ecotropic, amphotropic, polytropic, and xenotropic murine leukemia viruses (MLVs), indicating that infections by these viruses occur through acidic endosomes and require cathepsin proteases in the susceptible cells such as TE671 cells. However, as previously shown, the endosome acidification inhibitors did not inhibit these viral infections in XC cells. It is generally accepted that the ecotropic MLV infection in XC cells occurs at the plasma membrane. Because cathepsin proteases are activated by low pH in acidic endosomes, the acidification inhibitors may inhibit the viral infections by suppressing cathepsin protease activation. The acidification inhibitors attenuated the activities of cathepsin proteases B and L in TE671 cells, but not in XC cells. Processing of cathepsin protease L was suppressed by the acidification inhibitor in NIH3T3 cells, but again not in XC cells. These results indicate that cathepsin proteases are activated without endosome acidification in XC cells. Treatment with an endocytosis inhibitor or knockdown of dynamin 2 expression by siRNAs suppressed MLV infections in all examined cells including XC cells. Furthermore, endosomal cathepsin proteases were required for these viral infections in XC cells as other susceptible cells. These results suggest that infections of XC cells by the MLVs and Ebola virus occur through endosomes and pH-independent cathepsin activation induces pH-independent infection in XC cells.


Annals of the Rheumatic Diseases | 2014

AB0050 Irf8 Promotes TH17 Differentiation via Activation of Integrin-Mediated Tgfbeta Signaling in Neuroinflammation

Ryusuke Yoshimi; Yuko Yoshida; Hiroaki Yoshii; D. Kim; Anup Dey; Huabao Xiong; J. Munasinghe; Itaru Yazawa; M.J. O'Donovan; Olga A. Maximova; S. Sharma; J. Zhu; Hongsheng Wang; Herbert C. Morse; Yoshiaki Ishigatsubo; Keiko Ozato

Background There is accumulating evidence that Th17 cells promote inflammation and tissue injury in many autoimmune diseases. Although recent epidemiological studies identified IRF8 as a susceptibility factor for the autoimmune diseases, such as systemic lupus erythematosus1 and multiple sclerosis2, it has remained unknown how IRF8 influences the role of Th17 cell expansion in the pathogenesis. Objectives Here we investigated the role of IRF8 in Th17 differentiation using mice with experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. Methods Wild-type (WT), conventional Irf8 –/–, and two conditional Irf8 –/– mice, Irf8 f/f mice with LysM-cre and those with Lck-cre, were injected with MOG35–55 and clinical sign of the EAE were scored. The mRNA levels of cytokine and transcription factor genes and the populations of immune cell subsets in lymph nodes, spleens, and spinal cords were measured by qPCR and flow cytometry. Luciferase reporter assays and chromatin immunoprecipitation (ChIP) assays were performed to test IRF8 regulation of Itgb8 transcription. We examined in vitro development of Th17 cells from naïve CD4+ T cells cultured with CD11c+ dendritic cells (DCs) in the presence of latent TGFβ. Results Irf8 –/– mice were resistant to EAE and failed to generate Th1, Th17 and Treg cells after MOG injection. Irf8 f/f mice with LysM-cre remained resistant to EAE, while those with Lck-cre showed similar clinical scores as WT mice, indicating that IRF8 in APCs, but not in T cells, is responsible for causing EAE. The levels of Il12b, a subunit of IL-12p70 and IL-23, greatly increased in WT mice, but not in Irf8 –/– mice, during EAE. The increase in IL-27 production in Irf8 –/– mice was observed, indicating that IRF8 creates IL-12 family cytokine milieu favoring EAE. On the other hand, Itgb8, an integrin molecule important for Th17 differentiation via TGFβ activation, was highly expressed and in WT APCs, but was much lower in the Irf8 –/– counterparts. The reporter containing Itgb8 upstream fragment gave robust luciferase activity when co-transfected with Irf8 vector. Luciferase assays and ChIP assays showed that Itgb8 is a direct target of IRF8 which activates Itgb8 transcription. With latent TGFβ, WT DCs, but not Irf8 –/– DCs, stimulated Th17 differentiation. Conclusions This study shows that IRF8 is essential for Th17 cell development by controlling the production of IL-12 family cytokines and by activating TGFβ signaling in APCs. References BIOLUPUS Network, GENLES Network. Identification of IRF8, TMEM39A, and IKZF3-ZPBP2 as susceptibility loci for systemic lupus erythematosus in a large-scale multiracial replication study. Am J Hum Genet 2012;90:648-60. International Multiple Sclerosis Genetics Consortium (IMSGC). Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis. Nat Genet 2013;45:1353-60. Disclosure of Interest None declared DOI 10.1136/annrheumdis-2014-eular.3457


Virology | 2004

Determinant for the inhibition of ecotropic murine leukemia virus infection by N-linked glycosylation of the rat receptor

Yoshinao Kubo; Akinori Ishimoto; Takeshi Ono; Hiroaki Yoshii; Chika Tominaga; Chiho Mitani; Hiroshi Amanuma; Naoki Yamamoto

Collaboration


Dive into the Hiroaki Yoshii's collaboration.

Top Co-Authors

Avatar

Yoshinao Kubo

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yuetsu Tanaka

University of the Ryukyus

View shared research outputs
Top Co-Authors

Avatar

Herbert C. Morse

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Hongsheng Wang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Huabao Xiong

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Keiko Ozato

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge