Network


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

Hotspot


Dive into the research topics where Takahito Sanada is active.

Publication


Featured researches published by Takahito Sanada.


Journal of Experimental Medicine | 2006

IFNγ-dependent, spontaneous development of colorectal carcinomas in SOCS1-deficient mice

Toshikatsu Hanada; Takashi Kobayashi; Takatoshi Chinen; Kazuko Saeki; Hiromi Takaki; Keiko Koga; Yasumasa Minoda; Takahito Sanada; Tomoko Yoshioka; Hiromitsu Mimata; Seiya Kato; Akihiko Yoshimura

Approximately 20% of human cancers are estimated to develop from chronic inflammation. Recently, the NF-κB pathway was shown to play an essential role in promoting inflammation-associated cancer, but the role of the JAK/STAT pathway, another important signaling pathway of proinflammatory cytokines, remains to be investigated. Suppressor of cytokine signaling-1 (SOCS1) acts as an important physiological regulator of cytokine responses, and silencing of the SOCS1 gene by DNA methylation has been found in several human cancers. Here, we demonstrated that SOCS1-deficient mice (SOCS1−/−Tg mice), in which SOCS1 expression was restored in T and B cells on a SOCS1−/− background, spontaneously developed colorectal carcinomas carrying nuclear β-catenin accumulation and p53 mutations at 6 months of age. However, interferon (IFN)γ−/−SOCS1−/− mice and SOCS1−/−Tg mice treated with anti-IFNγ antibody did not develop such tumors. STAT3 and NF-κB activation was evident in SOCS1−/−Tg mice, but these were not sufficient for tumor development because these are also activated in IFNγ−/−SOCS1−/− mice. However, colons of SOCS1−/−Tg mice, but not IFNγ−/−SOCS1−/− mice, showed hyperactivation of STAT1, which resulted in the induction of carcinogenesis-related enzymes, cyclooxygenase-2 and inducible nitric oxide synthase. These data strongly suggest that SOCS1 is a unique antioncogene which prevents chronic inflammation-mediated carcinogenesis by regulation of the IFNγ/STAT1 pathways.


Nature | 2012

The Shigella flexneri effector OspI deamidates UBC13 to dampen the inflammatory response

Takahito Sanada; Minsoo Kim; Hitomi Mimuro; Masato Suzuki; Michinaga Ogawa; Akiho Oyama; Hiroshi Ashida; Taira Kobayashi; Tomohiro Koyama; Shinya Nagai; Yuri Shibata; Jin Gohda; Jun-ichiro Inoue; Tsunehiro Mizushima; Chihiro Sasakawa

Many bacterial pathogens can enter various host cells and then survive intracellularly, transiently evade humoral immunity, and further disseminate to other cells and tissues. When bacteria enter host cells and replicate intracellularly, the host cells sense the invading bacteria as damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) by way of various pattern recognition receptors. As a result, the host cells induce alarm signals that activate the innate immune system. Therefore, bacteria must modulate host inflammatory signalling and dampen these alarm signals. How pathogens do this after invading epithelial cells remains unclear, however. Here we show that OspI, a Shigella flexneri effector encoded by ORF169b on the large plasmid and delivered by the type ΙΙΙ secretion system, dampens acute inflammatory responses during bacterial invasion by suppressing the tumour-necrosis factor (TNF)-receptor-associated factor 6 (TRAF6)-mediated signalling pathway. OspI is a glutamine deamidase that selectively deamidates the glutamine residue at position 100 in UBC13 to a glutamic acid residue. Consequently, the E2 ubiquitin-conjugating activity required for TRAF6 activation is inhibited, allowing S. flexneri OspI to modulate the diacylglycerol–CBM (CARD–BCL10–MALT1) complex–TRAF6–nuclear-factor-κB signalling pathway. We determined the 2.0 Å crystal structure of OspI, which contains a putative cysteine–histidine–aspartic acid catalytic triad. A mutational analysis showed this catalytic triad to be essential for the deamidation of UBC13. Our results suggest that S. flexneri inhibits acute inflammatory responses in the initial stage of infection by targeting the UBC13–TRAF6 complex.


Cell Host & Microbe | 2013

The Shigella OspC3 Effector Inhibits Caspase-4, Antagonizes Inflammatory Cell Death, and Promotes Epithelial Infection

Taira Kobayashi; Michinaga Ogawa; Takahito Sanada; Hitomi Mimuro; Minsoo Kim; Hiroshi Ashida; Reiko Akakura; Mitsutaka Yoshida; Magdalena Kawalec; Jean-Marc Reichhart; Tsunehiro Mizushima; Chihiro Sasakawa

Caspase-mediated inflammatory cell death acts as an intrinsic defense mechanism against infection. Bacterial pathogens deploy countermeasures against inflammatory cell death, but the mechanisms by which they do this remain largely unclear. In a screen for Shigella flexneri effectors that regulate cell death during infection, we discovered that Shigella infection induced acute inflammatory, caspase-4-dependent epithelial cell death, which is counteracted by the bacterial OspC3 effector. OspC3 interacts with the caspase-4-p19 subunit and inhibits its activation by preventing caspase-4-p19 and caspase-4-p10 heterodimerization by depositing the conserved OspC3 X1-Y-X₂-D-X₃ motif at the putative catalytic pocket of caspase-4. Infection of guinea pigs with a Shigella ospC3-deficient mutant resulted in enhanced inflammatory cell death and associated symptoms, correlating with decreased bacterial burdens. Salmonella Typhimurium and enteropathogenic Escherichia coli infection also induced caspase-4-dependent epithelial death. These findings highlight the importance of caspase-4-dependent innate immune responses and demonstrate that Shigella delivers a caspase-4-specific inhibitor to delay epithelial cell death and promote infection.


Nature Communications | 2014

Epigenetic silencing of miR-210 increases the proliferation of gastric epithelium during chronic Helicobacter pylori infection

Kotaro Kiga; Hitomi Mimuro; Masato Suzuki; Aya Shinozaki-Ushiku; Taira Kobayashi; Takahito Sanada; Minsoo Kim; Michinaga Ogawa; Yuka W. Iwasaki; Hiroyuki Kayo; Yoko Fukuda-Yuzawa; Masakazu Yashiro; Masashi Fukayama; Taro Fukao; Chihiro Sasakawa

Persistent colonization of the gastric mucosa by Helicobacter pylori (Hp) elicits chronic inflammation and aberrant epithelial cell proliferation, which increases the risk of gastric cancer. Here we examine the ability of microRNAs to modulate gastric cell proliferation in response to persistent Hp infection and find that epigenetic silencing of miR-210 plays a key role in gastric disease progression. Importantly, DNA methylation of the miR-210 gene is increased in Hp-positive human gastric biopsies as compared with Hp-negative controls. Moreover, silencing of miR-210 in gastric epithelial cells promotes proliferation. We identify STMN1 and DIMT1 as miR-210 target genes and demonstrate that inhibition of miR-210 expression augments cell proliferation by activating STMN1 and DIMT1 . Together, our results highlight inflammation-induced epigenetic silencing of miR-210 as a mechanism of induction of chronic gastric diseases, including cancer, during Hp infection.


Current Opinion in Immunology | 2011

Shigella are versatile mucosal pathogens that circumvent the host innate immune system.

Hiroshi Ashida; Michinaga Ogawa; Hitomi Mimuro; Taira Kobayashi; Takahito Sanada; Chihiro Sasakawa

The intestinal mucosa is equipped with multiple innate immune defense systems that sense bacterial infection, transmit alarm signals to the immune system, defeat intruding bacteria, and renew damaged and aging epithelial cells. Nevertheless, mucosal bacterial pathogens have versatile pathogenic mechanisms that modulate the host inflammatory and immune responses, manipulate host cell death and survival signal pathways, and renovate the injured epithelium. These properties enable pathogens to adapt to the intestinal mucosal environment, exploit cellular and immune functions, and facilitate infection. Here we review current topics on host defense mechanisms against bacterial infection and the countermeasures that Shigella use to evade the innate immune system.


Journal of Biological Chemistry | 2005

FLN29, a Novel Interferon- and LPS-inducible Gene Acting as a Negative Regulator of Toll-like Receptor Signaling *

Ryuichi Mashima; Kazuko Saeki; Daisuke Aki; Yasumasa Minoda; Hiromi Takaki; Takahito Sanada; Takashi Kobayashi; Hiroyuki Aburatani; Yuji Yamanashi; Akihiko Yoshimura

Lipopolysaccharide (LPS) activates macrophages through toll-like receptor (TLR) 4. Although the mechanism of the TLR signaling pathway has been well documented, the mechanism of the negative regulation in response to LPS, particularly LPS tolerance, is still poorly understood. In this study we identified and characterized a novel interferon- and LPS-inducible gene, FLN29, which contains a TRAF6-related zinc finger motif and TRAF family member-associated NF-κB activator-related sequences. The induction of FLN29 was dependent on STAT1. The forced expression of FLN29 in macrophage-like RAW cells resulted in the suppression of TLR-mediated NF-κB and mitogen-activated protein kinase activation, while a reduced expression of FLN29 by small interfering RNA partly cancelled the down-regulation of LPS signaling. Furthermore, we demonstrated that NF-κB activation induced by TRAF6 and TAB2 was impaired by co-expression of FLN29, suggesting FLN29 may regulate the downstream of TRAF6. Taken together, FLN29 is a new negative feedback regulator of TLR signaling.


Current Opinion in Microbiology | 2011

Shigella deploy multiple countermeasures against host innate immune responses.

Hiroshi Ashida; Michinaga Ogawa; Minsoo Kim; Shiho Suzuki; Takahito Sanada; Claire Punginelli; Hitomi Mimuro; Chihiro Sasakawa

Although the intestinal epithelium is equipped with multiple defense systems that sense bacterial components, transmit alarms to the immune system, clear the bacteria, and renew the injured epithelial lining, mucosal bacterial pathogens are capable of efficiently colonizing the intestinal epithelium, because they have evolved systems that modulate the inflammatory and immune responses of the host and exploit the harmful environments as replicative niches. In this review we highlight current topics concerning Shigellas tactics that interfere with the innate immune systems.


Journal of Biological Chemistry | 2008

FLN29 deficiency reveals its negative regulatory role in the toll-like receptor (TLR) and retinoic acid-inducible gene I (RIG-I)-like helicase signaling pathway

Takahito Sanada; Giichi Takaesu; Ryuichi Mashima; Ryoko Yoshida; Takashi Kobayashi; Akihiko Yoshimura

FLN29 was identified as an interferon (IFN)-inducible gene, and it has been shown to suppress Toll-like receptor 4-mediated NF-κB activation by binding to TRAF6. To elucidate the physiological roles of FLN29, we generated FLN29-deficient mice. FLN29 deficiency resulted in hyper-response to LPS both in vivo and in vitro, demonstrating the negative regulatory role of FLN29 in TLR4 signaling. Furthermore, we found that FLN29–/– mice exhibited increased susceptibility to poly(I:C)-induced septic shock compared with WT mice. FLN29–/– fibroblasts were highly resistant to vesicular stomatitis virus infection, and these cells produced more IFN-β than WT cells did in response to not only intracellular poly(I:C) but also overexpression of IPS-1. Forced expression of FLN29 inhibited the IPS-1-dependent activation of both NF-κB and IRF3. We also found that FLN29 could interact with TRIF, IPS-1, TRAF3, and TRAF6. Together, these results suggest that FLN29, in addition to playing a negative regulatory role in the TLR4 signaling pathway, negatively regulates the RIG-I-like helicase signaling pathway at the level of IPS-1/TRAF6 and IPS-1/TRAF3 complexes.


Journal of Biological Chemistry | 2011

Attenuated CagA Oncoprotein in Helicobacter pylori from Amerindians in Peruvian Amazon

Masato Suzuki; Kotaro Kiga; Dangeruta Kersulyte; Jaime Cok; Catherine C. Hooper; Hitomi Mimuro; Takahito Sanada; Shiho Suzuki; Masaaki Oyama; Hiroko Kozuka-Hata; Shigeru Kamiya; Quan Ming Zou; Robert H. Gilman; Douglas E. Berg; Chihiro Sasakawa

Population genetic analyses of bacterial genes whose products interact with host tissues can give new understanding of infection and disease processes. Here we show that strains of the genetically diverse gastric pathogen Helicobacter pylori from Amerindians from the remote Peruvian Amazon contain novel alleles of cagA, a major virulence gene, and reveal distinctive properties of their encoded CagA proteins. CagA is injected into the gastric epithelium where it hijacks pleiotropic signaling pathways, helps Hp exploit its special gastric mucosal niche, and affects the risk that infection will result in overt gastroduodenal diseases including gastric cancer. The Amerindian CagA proteins contain unusual but functional tyrosine phosphorylation motifs and attenuated CRPIA motifs, which affect gastric epithelial proliferation, inflammation, and bacterial pathogenesis. Amerindian CagA proteins induced less production of IL-8 and cancer-associated Mucin 2 than did those of prototype Western or East Asian strains and behaved as dominant negative inhibitors of action of prototype CagA during mixed infection of Mongolian gerbils. We suggest that Amerindian cagA is of relatively low virulence, that this may have been selected in ancestral strains during infection of the people who migrated from Asia into the Americas many thousands of years ago, and that such attenuated CagA proteins could be useful therapeutically.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Shigella IpaH7.8 E3 ubiquitin ligase targets glomulin and activates inflammasomes to demolish macrophages

Shiho Suzuki; Hitomi Mimuro; Minsoo Kim; Michinaga Ogawa; Hiroshi Ashida; Takahito Toyotome; Luigi Franchi; Masato Suzuki; Takahito Sanada; Toshihiko Suzuki; Hiroko Tsutsui; Gabriel Núñez; Chihiro Sasakawa

Significance Shigella modulates macrophage cell death by activating nucleotide-binding oligomerization domain–like receptor (NLR) inflammasome to secure its own dissemination. Here we report that Shigella invasion plasmid antigen H7.8 (IpaH7.8) plays a central role in inducing macrophage cell death via activation of NLR family pyrin domain-containing 3 and NLR family CARD domain-containing 4 inflammasomes in an IpaH7.8 enzyme 3 (E3) ligase-dependent manner. Importantly, an IpaH7.8-deficient mutant was unable to egress from macrophages efficiently, resulting in delayed bacterial multiplication. We identified glomulin—a member of the S-phase kinase-associated protein 1–F-box–like complex that originally was identified as a protein required for normal vascular development—as a target for IpaH7.8 E3 ligase-mediated polyubiquitination, which leads to NLR inflammasome activation. In vitro and in vivo studies confirmed that IpaH7.8-mediated glomulin degradation during Shigella infection activated NLR inflammasomes and promoted cell death. When nucleotide-binding oligomerization domain–like receptors (NLRs) sense cytosolic-invading bacteria, they induce the formation of inflammasomes and initiate an innate immune response. In quiescent cells, inflammasome activity is tightly regulated to prevent excess inflammation and cell death. Many bacterial pathogens provoke inflammasome activity and induce inflammatory responses, including cell death, by delivering type III secreted effectors, the rod component flagellin, and toxins. Recent studies indicated that Shigella deploy multiple mechanisms to stimulate NLR inflammasomes through type III secretion during infection. Here, we show that Shigella induces rapid macrophage cell death by delivering the invasion plasmid antigen H7.8 (IpaH7.8) enzyme 3 (E3) ubiquitin ligase effector via the type III secretion system, thereby activating the NLR family pyrin domain-containing 3 (NLRP3) and NLR family CARD domain-containing 4 (NLRC4) inflammasomes and caspase-1 and leading to macrophage cell death in an IpaH7.8 E3 ligase-dependent manner. Mice infected with Shigella possessing IpaH7.8, but not with Shigella possessing an IpaH7.8 E3 ligase-null mutant, exhibited enhanced bacterial multiplication. We defined glomulin/flagellar-associated protein 68 (GLMN) as an IpaH7.8 target involved in IpaH7.8 E3 ligase-dependent inflammasome activation. This protein originally was identified through its association with glomuvenous malformations and more recently was described as a member of a Cullin ring ligase inhibitor. Modifying GLMN levels through overexpression or knockdown led to reduced or augmented inflammasome activation, respectively. Macrophages stimulated with lipopolysaccharide/ATP induced GLMN puncta that localized with the active form of caspase-1. Macrophages from GLMN+/− mice were more responsive to inflammasome activation than those from GLMN+/+ mice. Together, these results highlight a unique bacterial adaptation that hijacks inflammasome activation via interactions between IpaH7.8 and GLMN.

Collaboration


Dive into the Takahito Sanada's collaboration.

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
Top Co-Authors

Avatar

Minsoo Kim

University of Rochester

View shared research outputs
Researchain Logo
Decentralizing Knowledge