Network


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

Hotspot


Dive into the research topics where Miho Watanabe-Takahashi is active.

Publication


Featured researches published by Miho Watanabe-Takahashi.


Journal of Cell Science | 2009

Yip1A regulates the COPI-independent retrograde transport from the Golgi complex to the ER

Fumi Kano; Shinobu Yamauchi; Yumi Yoshida; Miho Watanabe-Takahashi; Kiyotaka Nishikawa; Nobuhiro Nakamura; Masayuki Murata

Yip1A, a mammalian homologue of yeast Yip1p, is a multi-spanning membrane protein that is considered to be involved in transport between the endoplasmic reticulum (ER) and the Golgi. However, the precise role of Yip1A in mammalian cells remains unclear. We show here that endogenous Yip1A is localized to the ER-Golgi intermediate compartment (ERGIC). Knockdown of Yip1A by RNAi did not induce morphological changes in the Golgi, ER, or ERGIC. By analyzing a number of intracellular transport pathways, we found that Yip1A knockdown delayed the transport of Shiga toxin from the Golgi to the ER, but did not affect the anterograde transport of VSVGts045. We also found that a recombinant protein that corresponded to the N-terminal domain of Yip1A inhibited the COPI-independent retrograde transport of GFP-tagged galactosyltransferase, GT-GFP, but not the COPI-dependent retrograde transport of p58/ERGIC53. Furthermore, we found that Yip1A knockdown resulted in the dissociation of Rab6 from the membranes. These results suggested that Yip1A has a role in COPI-independent retrograde transport from the Golgi to the ER and regulates the membrane recruitment of Rab6.


Nature Communications | 2013

Substrate ectodomain is critical for substrate preference and inhibition of γ-secretase

Satoru Funamoto; Toru Sasaki; Seiko Ishihara; Mika Nobuhara; Masaki Nakano; Miho Watanabe-Takahashi; Takashi Saito; Nobuto Kakuda; Tomohiro Miyasaka; Kiyotaka Nishikawa; Takaomi C. Saido; Yasuo Ihara

Understanding the substrate recognition mechanism of γ-secretase is a key step for establishing substrate-specific inhibition of amyloid β-protein (Aβ) production. However, it is widely believed that γ-secretase is a promiscuous protease and that its substrate-specific inhibition is elusive. Here we show that γ-secretase distinguishes the ectodomain length of substrates and preferentially captures and cleaves substrates containing a short ectodomain. We also show that a subset of peptides containing the CDCYCxxxxCxCxSC motif binds to the amino terminus of C99 and inhibits Aβ production in a substrate-specific manner. Interestingly, these peptides suppress β-secretase-dependent cleavage of APP, but not that of sialyltransferase 1. Most importantly, intraperitoneal administration of peptides into mice results in a significant reduction in cerebral Aβ levels. This report provides direct evidence of the substrate preference of γ-secretase and its mechanism. Our results demonstrate that the ectodomain of C99 is a potent target for substrate-specific anti-Aβ therapeutics to combat Alzheimer’s disease.


Infection and Immunity | 2010

An Orally Applicable Shiga Toxin Neutralizer Functions in the Intestine To Inhibit the Intracellular Transport of the Toxin

Miho Watanabe-Takahashi; Toshio Sato; Taeko Dohi; Noriko Noguchi; Fumi Kano; Masayuki Murata; Takashi Hamabata; Yasuhiro Natori; Kiyotaka Nishikawa

ABSTRACT Shiga toxin 2 (Stx2) is a major virulence factor in infections with Stx-producing Escherichia coli (STEC), which causes gastrointestinal diseases and sometimes fatal systemic complications. Recently, we developed an oral Stx2 inhibitor known as Ac-PPP-tet that exhibits remarkable therapeutic potency in an STEC infection model. However, the precise mechanism underlying the in vivo therapeutic effects of Ac-PPP-tet is unknown. Here, we found that Ac-PPP-tet completely inhibited fluid accumulation in the rabbit ileum caused by the direct injection of Stx2. Interestingly, Ac-PPP-tet accumulated in the ileal epithelial cells only through its formation of a complex with Stx2. The formation of Ac-PPP-tet-Stx2 complexes in cultured epithelial cells blocked the intracellular transport of Stx2 from the Golgi apparatus to the endoplasmic reticulum, a process that is essential for Stx2 cytotoxicity. Thus, Ac-PPP-tet is the first Stx neutralizer that functions in the intestine by altering the intracellular transport of Stx2 in epithelial cells.


Infection and Immunity | 2013

Identification of a peptide-based neutralizer that potently inhibits both Shiga toxins 1 and 2 by targeting specific receptor-binding regions.

Kazue Tsutsuki; Miho Watanabe-Takahashi; Yasuaki Takenaka; Eiji Kita; Kiyotaka Nishikawa

ABSTRACT Shiga toxin (Stx) is a major virulence factor of enterohemorrhagic Escherichia coli that occasionally causes fatal systemic complications. We recently developed a tetravalent peptide (PPP-tet) that neutralizes the cytotoxicity of Stx2 using a multivalent peptide library approach. In this study, we used this technique to identify a series of tetravalent peptides that bound to Stx1, another major Stx family member, with high affinity by targeting one receptor-binding site of the B subunit. One peptide, MMA-tet, markedly inhibited Stx1 and Stx2 cytotoxicity with greater potency than PPP-tet. After forming a complex with Stx1 through its specific receptor-binding region, MMA-tet did not affect vesicular transport of the toxin to the endoplasmic reticulum but substantially rescued inhibition of the protein synthesis induced by Stx1. Oral application of MMA-tet protected mice from a fatal dose of an E. coli O157:H7 strain producing both toxins. MMA-tet may be a promising therapeutic agent against the infection.


FEBS Open Bio | 2015

Proteasome inhibitors prevent cell death and prolong survival of mice challenged by Shiga toxin

Takayuki Hattori; Miho Watanabe-Takahashi; Nobumichi Ohoka; Takashi Hamabata; Koichi Furukawa; Kiyotaka Nishikawa; Mikihiko Naito

Shiga toxin (Stx) causes fatal systemic complications. Stx induces apoptosis, but the mechanism of which is unclear. We report that Stx induced rapid reduction of short‐lived anti‐apoptotic proteins followed by activation of caspase 9 and the progression of apoptosis. Proteasome inhibitors prevented the reduction of anti‐apoptotic proteins, and inhibited caspase activation and apoptosis, suggesting that the reduction of anti‐apoptotic proteins is a prerequisite for Stx‐induced apoptosis. A clinically approved proteasome inhibitor, bortezomib, prolonged the survival of mice challenged by Stx. These results imply that proteasome inhibition may be a novel approach to prevent the fatal effects of Stx.


PLOS ONE | 2015

Development of a Novel Tetravalent Synthetic Peptide That Binds to Phosphatidic Acid

Rina Ogawa; Kohjiro Nagao; Kentaro Taniuchi; Masaki Tsuchiya; Utako Kato; Yuji Hara; Takehiko Inaba; Toshihide Kobayashi; Yoshihiro Sasaki; Kazunari Akiyoshi; Miho Watanabe-Takahashi; Kiyotaka Nishikawa; Masato Umeda

We employed a multivalent peptide-library screening technique to identify a peptide motif that binds to phosphatidic acid (PA), but not to other phospholipids such as phosphatidylcholine (PC), phosphatidylethanolamine (PE), and phosphatidylserine (PS). A tetravalent peptide with the sequence motif of MARWHRHHH, designated as PAB-TP (phosphatidic acid-binding tetravalent peptide), was shown to bind as low as 1 mol% of PA in the bilayer membrane composed of PC and cholesterol. Kinetic analysis of the interaction between PAB-TP and the membranes containing 10 mol% of PA showed that PAB-TP associated with PA with a low dissociation constant of KD = 38 ± 5 nM. Coexistence of cholesterol or PE with PA in the membrane enhanced the PAB-TP binding to PA by increasing the ionization of the phosphomonoester head group as well as by changing the microenvironment of PA molecules in the membrane. Amino acid replacement analysis demonstrated that the tryptophan residue at position 4 of PAB-TP was involved in the interaction with PA. Furthermore, a series of amino acid substitutions at positions 5 to 9 of PAB-TP revealed the involvement of consecutive histidine and arginine residues in recognition of the phosphomonoester head group of PA. Our results demonstrate that the recognition of PA by PAB-TP is achieved by a combination of hydrophobic, electrostatic and hydrogen-bond interactions, and that the tetravalent structure of PAB-TP contributes to the high affinity binding to PA in the membrane. The novel PA-binding tetravalent peptide PAB-TP will provide insight into the molecular mechanism underlying the recognition of PA by PA-binding proteins that are involved in various cellular events.


Applied and Environmental Microbiology | 2015

Identification of a Wide Range of Motifs Inhibitory to Shiga Toxin by Affinity-Driven Screening of Customized Divalent Peptides Synthesized on a Membrane

Mihoko Kato; Miho Watanabe-Takahashi; Eiko Shimizu; Kiyotaka Nishikawa

ABSTRACT Shiga toxin (Stx), a major virulence factor of enterohemorrhagic Escherichia coli, binds to target cells through a multivalent interaction between its B-subunit pentamer and the cell surface receptor globotriaosylceramide, resulting in a remarkable increase in its binding affinity. This phenomenon is referred to as the “clustering effect.” Previously, we developed a multivalent peptide library that can exert the clustering effect and identified Stx neutralizers with tetravalent peptides by screening this library for high-affinity binding to the specific receptor-binding site of the B subunit. However, this technique yielded only a limited number of binding motifs, with some redundancy in amino acid selectivity. In this study, we established a novel technique to synthesize up to 384 divalent peptides whose structures were customized to exert the clustering effect on the B subunit on a single cellulose membrane. By targeting Stx1a, a major Stx subtype, the customized divalent peptides were screened to identify high-affinity binding motifs. The sequences of the peptides were designed based on information obtained from the multivalent peptide library technique. A total of 64 candidate motifs were successfully identified, and 11 of these were selected to synthesize tetravalent forms of the peptides. All of the synthesized tetravalent peptides bound to the B subunit with high affinities and effectively inhibited the cytotoxicity of Stx1a in Vero cells. Thus, the combination of the two techniques results in greatly improved efficiency in identifying biologically active neutralizers of Stx.


Genes to Cells | 2016

M-COPA, a novel Golgi system disruptor, suppresses apoptosis induced by Shiga toxin

Takayuki Hattori; Miho Watanabe-Takahashi; Isamu Shiina; Y. Ohashi; Shingo Dan; Kiyotaka Nishikawa; Takao Yamori; Mikihiko Naito

Shiga toxin (Stx) is a main virulence factor of Stx‐producing Escherichia coli (STEC) that contributes to diarrhea and hemorrhagic colitis and occasionally to fatal systemic complications. Therefore, the development of an antidote to neutralize Stx toxicity is urgently needed. After internalization into cells, Stx is transferred to the Golgi apparatus via a retrograde vesicular transport system. We report here that 2‐methylcoprophilinamide (M‐COPA), a compound that induces disassembly of the Golgi apparatus by inactivating ADP‐ribosylation factor 1 (Arf1), suppresses Stx‐induced apoptosis. M‐COPA inhibited transport of Stx from the plasma membrane to the Golgi apparatus and suppressed degradation of anti‐apoptotic proteins and the activation of caspases. These findings suggest that inhibition of Stx retrograde transport by M‐COPA could be a novel approach to suppress Stx toxicity.


Scientific Reports | 2018

Exosome-associated Shiga toxin 2 is released from cells and causes severe toxicity in mice

Miho Watanabe-Takahashi; Shinji Yamasaki; Masayuki Murata; Fumi Kano; Jun Motoyama; Jyoji Yamate; Jumpei Omi; Waka Sato; Hirofumi Ukai; Kentaro Shimasaki; Masaya Ikegawa; Miwa Tamura-Nakano; Ryohei Yanoshita; Yuri Nishino; Atsuo Miyazawa; Yasuhiro Natori; Noriko Toyama-Sorimachi; Kiyotaka Nishikawa

Shiga toxin (Stx), a major virulence factor of enterohemorrhagic Escherichia coli (EHEC), is classified into two subgroups, Stx1 and Stx2. Clinical data clearly indicate that Stx2 is associated with more severe toxicity than Stx1, but the molecular mechanism underlying this difference is not fully understood. Here, we found that after being incorporated into target cells, Stx2, can be transported by recycling endosomes, as well as via the regular retrograde transport pathway. However, transport via recycling endosome did not occur with Stx1. We also found that Stx2 is actively released from cells in a receptor-recognizing B-subunit dependent manner. Part of the released Stx2 is associated with microvesicles, including exosome markers (referred to as exo-Stx2), whose origin is in the multivesicular bodies that formed from late/recycling endosomes. Finally, intravenous administration of exo-Stx2 to mice causes more lethality and tissue damage, especially severe renal dysfunction and tubular epithelial cell damage, compared to a free form of Stx2. Thus, the formation of exo-Stx2 might contribute to the severity of Stx2 in vivo, suggesting new therapeutic strategies against EHEC infections.


Genes to Cells | 2018

Pleckstrin homology domain of p210 BCR‐ABL interacts with cardiolipin to regulate its mitochondrial translocation and subsequent mitophagy

Kentaro Shimasaki; Miho Watanabe-Takahashi; Masato Umeda; Satoru Funamoto; Yoshiro Saito; Noriko Noguchi; Keigo Kumagai; Kentaro Hanada; Fujiko Tsukahara; Yoshiro Maru; Norihito Shibata; Mikihiko Naito; Kiyotaka Nishikawa

Chronic myeloid leukemia (CML) is caused by the chimeric protein p210 BCR‐ABL encoded by a gene on the Philadelphia chromosome. Although the kinase domain of p210 BCR‐ABL is an active driver of CML, the pathological role of its pleckstrin homology (PH) domain remains unclear. Here, we carried out phospholipid vesicle‐binding assays to show that cardiolipin (CL), a characteristic mitochondrial phospholipid, is a unique ligand of the PH domain. Arg726, a basic amino acid in the ligand‐binding region, was crucial for ligand recognition. A subset of wild‐type p210 BCR‐ABL that was transiently expressed in HEK293 cells was dramatically translocated from the cytosol to mitochondria in response to carbonyl cyanide m‐chlorophenylhydrazone (CCCP) treatment, which induces mitochondrial depolarization and subsequent externalization of CL to the organelles outer membrane, whereas an R726A mutant of the protein was not translocated. Furthermore, only wild‐type p210 BCR‐ABL, but not the R726A mutant, suppressed CCCP‐induced mitophagy and subsequently enhanced reactive oxygen species production. Thus, p210 BCR‐ABL can change its intracellular localization via interactions between the PH domain and CL to cope with mitochondrial damage. This suggests that p210 BCR‐ABL could have beneficial effects for cancer proliferation, providing new insight into the PH domains contribution to CML pathogenesis.

Collaboration


Dive into the Miho Watanabe-Takahashi'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
Researchain Logo
Decentralizing Knowledge