72 results on '"Kiyotaka Nishikawa"'
Search Results
2. Hepatitis B Virus Utilizes a Retrograde Trafficking Route via the Trans-Golgi Network to Avoid Lysosomal DegradationSummary
- Author
-
Ying-Yi Li, Kazuyuki Kuroki, Tetsuro Shimakami, Kazuhisa Murai, Kazunori Kawaguchi, Takayoshi Shirasaki, Kouki Nio, Saiho Sugimoto, Tomoki Nishikawa, Hikari Okada, Noriaki Orita, Hideo Takayama, Ying Wang, Phuong Doan Thi Bich, Astuya Ishida, Sadahiro Iwabuchi, Shinichi Hashimoto, Takeshi Shimaoka, Noriko Tabata, Miho Watanabe-Takahashi, Kiyotaka Nishikawa, Hiroshi Yanagawa, Motoharu Seiki, Kouji Matsushima, Taro Yamashita, Shuichi Kaneko, and Masao Honda
- Subjects
cccDNA ,DOCK11 ,Retrograde Trafficking ,AGAP2 ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Background & Aims: Hepatitis B virus (HBV) infection is difficult to cure owing to the persistence of covalently closed circular viral DNA (cccDNA). We performed single-cell transcriptome analysis of newly established HBV-positive and HBV-negative hepatocellular carcinoma cell lines and found that dedicator of cytokinesis 11 (DOCK11) was crucially involved in HBV persistence. However, the roles of DOCK11 in the HBV lifecycle have not been clarified. Methods: The cccDNA levels were measured by Southern blotting and real-time detection polymerase chain reaction in various hepatocytes including PXB cells by using an HBV-infected model. The retrograde trafficking route of HBV capsid was investigated by super-resolution microscopy, proximity ligation assay, and time-lapse analysis. The downstream molecules of DOCK11 and underlying mechanism were examined by liquid chromatography-tandem mass spectrometry, immunoblotting, and enzyme-linked immunosorbent assay. Results: The cccDNA levels were strongly increased by DOCK11 overexpression and repressed by DOCK11 suppression. Interestingly, DOCK11 functionally associated with retrograde trafficking proteins in the trans-Golgi network (TGN), Arf-GAP with GTPase domain, ankyrin repeat, and pleckstrin homology domain-containing protein 2 (AGAP2), and ADP-ribosylation factor 1 (ARF1), together with HBV capsid, to open an alternative retrograde trafficking route for HBV from early endosomes (EEs) to the TGN and then to the endoplasmic reticulum (ER), thereby avoiding lysosomal degradation. Clinically, DOCK11 levels in liver biopsies from patients with chronic hepatitis B were significantly reduced by entecavir treatment, and this reduction correlated with HBV surface antigen levels. Conclusions: HBV uses a retrograde trafficking route via EEs-TGN-ER for infection that is facilitated by DOCK11 and serves to maintain cccDNA. Therefore, DOCK11 is a potential therapeutic target to prevent persistent HBV infection.
- Published
- 2023
- Full Text
- View/download PDF
3. A unique peptide-based pharmacophore identifies an inhibitory compound against the A-subunit of Shiga toxin
- Author
-
Miho Watanabe-Takahashi, Miki Senda, Ryunosuke Yoshino, Masahiro Hibino, Shinichiro Hama, Tohru Terada, Kentaro Shimizu, Toshiya Senda, and Kiyotaka Nishikawa
- Subjects
Medicine ,Science - Abstract
Abstract Shiga toxin (Stx), a major virulence factor of enterohemorrhagic Escherichia coli (EHEC), can cause fatal systemic complications. Recently, we identified a potent inhibitory peptide that binds to the catalytic A-subunit of Stx. Here, using biochemical structural analysis and X-ray crystallography, we determined a minimal essential peptide motif that occupies the catalytic cavity and is required for binding to the A-subunit of Stx2a, a highly virulent Stx subtype. Molecular dynamics simulations also identified the same motif and allowed determination of a unique pharmacophore for A-subunit binding. Notably, a series of synthetic peptides containing the motif efficiently inhibit Stx2a. In addition, pharmacophore screening and subsequent docking simulations ultimately identified nine Stx2a-interacting molecules out of a chemical compound database consisting of over 7,400,000 molecules. Critically, one of these molecules markedly inhibits Stx2a both in vitro and in vivo, clearly demonstrating the significance of the pharmacophore for identifying therapeutic agents against EHEC infection.
- Published
- 2022
- Full Text
- View/download PDF
4. Identification of a peptide motif that potently inhibits two functionally distinct subunits of Shiga toxin
- Author
-
Miho Watanabe-Takahashi, Masakazu Tamada, Miki Senda, Masahiro Hibino, Eiko Shimizu, Akiko Okuta, Atsuo Miyazawa, Toshiya Senda, and Kiyotaka Nishikawa
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Watanabe-Takahashi, Tamada, Senda et al. identify a tetravalent peptide that inhibits Shiga toxin (Stx), a major virulence factor of enterohemorrhagic Escherichia coli, by targeting its receptor-binding. On the other hand, a monomeric peptide containing the same motif occupies the Stx catalytic cavity, suggesting that this peptide motif can inhibit two subunits of Stx.
- Published
- 2021
- Full Text
- View/download PDF
5. The inducible amphisome isolates viral hemagglutinin and defends against influenza A virus infection
- Author
-
Jumpei Omi, Miho Watanabe-Takahashi, Katsura Igai, Eiko Shimizu, Ching-Yi Tseng, Tomohiro Miyasaka, Tsuyoshi Waku, Shinichiro Hama, Rieka Nakanishi, Yuki Goto, Yuri Nishino, Atsuo Miyazawa, Yasuhiro Natori, Makoto Yamashita, and Kiyotaka Nishikawa
- Subjects
Science - Abstract
The influenza virus hemagglutinin (HA) protein is a target for antivirals. Here, using a multivalent peptide library screen, the authors identify PVF-tet as an HA inhibitor and show that the peptide inhibits IAV replication by sequestering HA into amphisomes, suggesting these to represent a type of anti-viral machinery.
- Published
- 2020
- Full Text
- View/download PDF
6. Proteasome inhibitors prevent cell death and prolong survival of mice challenged by Shiga toxin
- Author
-
Takayuki Hattori, Miho Watanabe-Takahashi, Nobumichi Ohoka, Takashi Hamabata, Koichi Furukawa, Kiyotaka Nishikawa, and Mikihiko Naito
- Subjects
Shiga toxin ,Apoptosis ,Apoptosis inhibitory proteins ,Proteasome ,Proteasome inhibitor ,Biology (General) ,QH301-705.5 - Abstract
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.
- Published
- 2015
- Full Text
- View/download PDF
7. Development of a Novel Tetravalent Synthetic Peptide That Binds to Phosphatidic Acid.
- Author
-
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, and Masato Umeda
- Subjects
Medicine ,Science - Abstract
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.
- Published
- 2015
- Full Text
- View/download PDF
8. Novel multivalent S100A8 inhibitory peptides attenuate tumor progression and metastasis by inhibiting the TLR4-dependent pathway
- Author
-
Atsuko Deguchi, Miho Watanabe-Takahashi, Taishi Mishima, Tsutomu Omori, Umeharu Ohto, Nobuto Arashiki, Fumio Nakamura, Kiyotaka Nishikawa, and Yoshiro Maru
- Subjects
Cancer Research ,Molecular Medicine ,Molecular Biology - Abstract
The tumor-elicited inflammation is closely related to tumor microenvironment during tumor progression. S100A8, an endogenous ligand of Toll-like receptor 4 (TLR4), is known as a key molecule in the tumor microenvironment and premetastatic niche formation. We firstly generated a novel multivalent S100A8 competitive inhibitory peptide (divalent peptide3A5) against TLR4/MD-2, using the alanine scanning. Divalent peptide3A5 suppressed S100A8-mediated interleukin-8 and vascular endothelial growth factor production in human colorectal tumor SW480 cells. Using SW480-transplanted xenograft models, divalent peptide3A5 suppressed tumor progression in a dose-dependent manner. We demonstrated that combination therapy with divalent peptide3A5 and bevacizumab synergistically suppressed tumor growth in SW480 xenograft models. Using syngeneic mouse models, we found that divalent peptide3A5 improved the efficacy of anti-programmed death (PD)1 antibody, and lung metastasis. In addition, by using multivalent peptide library screening based on peptide3A5, we then isolated two more candidates; divalent ILVIK, and tetravalent ILVIK. Of note, multivalent ILVIK, but not monovalent ILVIK showed competitive inhibitory activity against TLR4/MD-2 complex, and anti-tumoral activity in SW480 xenograft models. As most tumor cells including SW480 cells also express TLR4, S100A8 inhibitory peptides would target both the tumor microenvironment and tumor cells. Thus, multivalent S100A8 inhibitory peptides would provide new pharmaceutical options for aggressive cancers.
- Published
- 2023
- Full Text
- View/download PDF
9. A tetravalent peptide that binds to the RANK-binding region of TRAF6 via a multivalent interaction efficiently inhibits osteoclast differentiation
- Author
-
Masataka Anzai, Miho Watanabe-Takahashi, Hiroshi Kawabata, Saori Mizuno, Yuu Taguchi, Jun-ichiro Inoue, and Kiyotaka Nishikawa
- Subjects
TNF Receptor-Associated Factor 6 ,Receptor Activator of Nuclear Factor-kappa B ,RANK Ligand ,Biophysics ,NF-kappa B ,Osteoclasts ,Cell Differentiation ,Cell Biology ,Peptides ,Molecular Biology ,Biochemistry - Abstract
Inhibition of osteoclast differentiation is a promising approach for the treatment of osteoporosis and rheumatoid arthritis. Receptor activator of nuclear factor kappa B (NF-κB) (RANK), which is an essential molecule for osteoclast differentiation, interacts with tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) to transduce downstream signals. Both RANK and TRAF6 have homo-trimeric structures, forming a multivalent interaction between the Pro-X-Glu-X-X-(aromatic/acidic) motif of RANK and the C-terminal domain of TRAF6 (TRAF-C), that markedly increases the binding affinity. Here, we designed a tetravalent peptide, RANK-tet, containing the TRAF-C-binding motif of RANK and found that RANK-tet binds to TRAF-C with high affinity. In contrast, a monomeric form of RANK-tet (RANK-mono) with the same TRAF-C-binding motif did not bind to TRAF-C, clearly indicating the multivalent interaction is strictly required for the high-affinity binding to TRAF-C. RANK-tet did not bind to a series of TRAF-C-mutants with an amino acid substitution in the RANK-binding region, indicating that RANK-tet specifically targets the RANK-binding region of TRAF-C. A cell-permeable form of RANK-tet that has poly-Arg residues at each C-terminal of the TRAF-C-binding motif efficiently inhibited the RANK ligand (RANKL)-induced differentiation of bone marrow cells to osteoclasts. Thus, this compound can be an effective anti-osteoclastogenic agent.
- Published
- 2022
10. Development of a novel tetravalent peptide that absorbs subtilase cytotoxin by targeting the receptor-binding B-subunit
- Author
-
Shinichiro Hama, Miki Nakahara, Miho Watanabe-Takahashi, Eiko Shimizu, Hiroyasu Tsutsuki, Kinnosuke Yahiro, and Kiyotaka Nishikawa
- Subjects
Shiga-Toxigenic Escherichia coli ,Cytotoxins ,Peptide Library ,Polymers ,Virulence Factors ,Escherichia coli Proteins ,Biophysics ,Cell Biology ,Subtilisins ,Carrier Proteins ,Cellulose ,Molecular Biology ,Biochemistry - Abstract
Subtilase cytotoxin (SubAB) is a major virulence factor produced by eae-negative Shiga-toxigenic Escherichia coli (STEC) that can cause fatal systemic complications. SubAB binds to target cells through multivalent interactions between its B-subunit pentamer and receptor molecules such as glycoproteins with a terminal N-glycolylneuraminic acid (Neu5Gc). We screened randomized multivalent peptide libraries synthesized on a cellulose membrane and identified a series of tetravalent peptides that efficiently bind to the receptor-binding region of the SubAB B-subunit pentamer. These peptides competitively inhibited the binding of the B-subunit to a receptor-mimic molecule containing clustered Neu5Gc (Neu5Gc-polymer). We selected the peptide with the highest inhibitory efficacy, FFP-tet, and covalently bound it to beads to synthesize FFP-tet-beads, a highly clustered SubAB absorber that displayed potency to absorb SubAB cytotoxicity through direct binding to the toxin. The efficacy of FFP-tet-beads to absorb SubAB cytotoxicity in solution was similar to that of Neu5Gc-polymer, suggesting that FFP-tet-beads might be an effective therapeutic agent against complications arising from eae-negative STEC infection.
- Published
- 2022
11. Novel multivalent S100A8 inhibitory peptides attenuate tumor progression and metastasis by inhibiting the TLR4-dependent pathway
- Author
-
Atsuko Deguchi, Miho Watanabe-Takahashi, Taishi Mishima, Tsutomu Omori, Umeharu Ohto, Kiyotaka Nishikawa, and Yoshiro Maru
- Abstract
The tumor-elicited inflammation is closely related to tumor microenvironment during tumor progression. S100A8, an endogenous ligand of Toll-like receptor 4 (TLR4), is known as a key molecule in the tumor microenvironment and premetastatic niche formation. We firstly generated a novel multivalent S100A8 competitive inhibitory peptide (divalent peptide3A5) against TLR4/MD-2, using the alanine scanning. Divalent peptide3A5 suppressed S100A8-mediated interleukin-8 and vascular endothelial growth factor production in human colorectal tumor SW480 cells. Using SW480-transplanted xenograft models, divalent peptide3A5 suppressed tumor progression in a dose-dependent manner. We demonstrated that combination therapy with divalent peptide3A5 and bevacizumab synergistically suppressed tumor growth in SW480 xenograft models. Using syngeneic mouse models, we found that divalent peptide3A5 improved the efficacy of anti-programmed death (PD)1 antibody, and lung metastasis. In addition, by using multivalent peptide library screening based on peptide3A5, we then isolated two more candidates; divalent ILVIK, and tetravalent ILVIK. Of note, multivalent ILVIK, but not monovalent ILVIK showed competitive inhibitory activity against TLR4/MD-2 complex, and anti-tumoral activity in SW480 xenograft models. As most tumor cells including SW480 cells also express TLR4, S100A8 inhibitory peptides would target both the tumor microenvironment and tumor cells. Thus, multivalent S100A8 inhibitory peptides would provide new pharmaceutical options for aggressive cancers.
- Published
- 2022
- Full Text
- View/download PDF
12. A nontoxigenic form of Shiga toxin 2 suppresses the production of amyloid β by altering the intracellular transport of amyloid precursor protein through its receptor-binding B-subunit
- Author
-
Satoru Funamoto, Koichi Furukawa, Waka Sato, Takashi Hamabata, Kiyotaka Nishikawa, and Miho Watanabe-Takahashi
- Subjects
0301 basic medicine ,Cell Survival ,Endosome ,Protein subunit ,Biophysics ,CHO Cells ,Endosomes ,Endocytosis ,Shiga Toxin 2 ,Biochemistry ,Amyloid beta-Protein Precursor ,03 medical and health sciences ,symbols.namesake ,Cricetulus ,0302 clinical medicine ,Catalytic Domain ,Extracellular ,Amyloid precursor protein ,Animals ,Humans ,Molecular Biology ,Amyloid beta-Peptides ,Globosides ,biology ,Chemistry ,Trihexosylceramides ,Endoplasmic reticulum ,Cell Membrane ,Shiga toxin ,Cell Biology ,Golgi apparatus ,Recombinant Proteins ,Cell biology ,Protein Transport ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Phosphatidylcholines ,symbols ,biology.protein ,Lysosomes - Abstract
Accumulation of amyloid-β peptide (Aβ) in neuronal cells and in the extracellular regions in the brain is a major cause of Alzheimer's disease (AD); therefore, inhibition of Aβ accumulation offers a promising approach for therapeutic strategies against AD. Aβ is produced by sequential proteolysis of amyloid precursor protein (APP) in late/recycling endosomes after endocytosis of APP located in the plasma membrane. Aβ is then released from cells in a free form or in an exosome-bound form. Shiga toxin (Stx) is a major virulence factor of enterohemorrhagic Escherichia coli. Recently, we found that one of the Stx subtypes, Stx2a, has a unique intracellular transport route after endocytosis through its receptor-binding B-subunit. A part of Stx2a can be transported to late/recycling endosomes and then degraded in a lysosomal acidic compartment, although in general Stx is transported to the Golgi and then to the endoplasmic reticulum in a retrograde manner. In this study, we found that treatment of APP-expressing cells with a mutant Stx2a (mStx2a), lacking cytotoxic activity because of mutations in the catalytic A-subunit, stimulated the transport of APP to the acidic compartment, which led to degradation of APP and a reduction in the amount of Aβ. mStx2a-treatment also inhibited the extracellular release of Aβ. Therefore, mStx2a may provide a new strategy to inhibit the production of Aβ by modulating the intracellular transport of APP.
- Published
- 2021
- Full Text
- View/download PDF
13. Identification of a peptide motif that potently inhibits two functionally distinct subunits of Shiga toxin
- Author
-
Miki Senda, Kiyotaka Nishikawa, Eiko Shimizu, Toshiya Senda, Masahiro Hibino, Miho Watanabe-Takahashi, Atsuo Miyazawa, Akiko Okuta, and Masakazu Tamada
- Subjects
0301 basic medicine ,Bacterial toxins ,Pentamer ,QH301-705.5 ,030106 microbiology ,Medicine (miscellaneous) ,Peptide ,medicine.disease_cause ,Crystallography, X-Ray ,General Biochemistry, Genetics and Molecular Biology ,Virulence factor ,Article ,Shiga Toxin ,03 medical and health sciences ,chemistry.chemical_compound ,Catalytic Domain ,hemic and lymphatic diseases ,Chlorocebus aethiops ,medicine ,Animals ,Biology (General) ,Cytotoxicity ,Escherichia coli ,Vero Cells ,X-ray crystallography ,Cell Proliferation ,chemistry.chemical_classification ,biology ,Shiga toxin ,Peptide Fragments ,030104 developmental biology ,Monomer ,Biochemistry ,chemistry ,biology.protein ,General Agricultural and Biological Sciences ,Protein Binding - Abstract
Shiga toxin (Stx) is a major virulence factor of enterohemorrhagic Escherichia coli, which causes fatal systemic complications. Here, we identified a tetravalent peptide that inhibited Stx by targeting its receptor-binding, B-subunit pentamer through a multivalent interaction. A monomeric peptide with the same motif, however, did not bind to the B-subunit pentamer. Instead, the monomer inhibited cytotoxicity with remarkable potency by binding to the catalytic A-subunit. An X-ray crystal structure analysis to 1.6 Å resolution revealed that the monomeric peptide fully occupied the catalytic cavity, interacting with Glu167 and Arg170, both of which are essential for catalytic activity. Thus, the peptide motif demonstrated potent inhibition of two functionally distinct subunits of Stx., Watanabe-Takahashi, Tamada, Senda et al. identify a tetravalent peptide that inhibits Shiga toxin (Stx), a major virulence factor of enterohemorrhagic Escherichia coli, by targeting its receptor-binding. On the other hand, a monomeric peptide containing the same motif occupies the Stx catalytic cavity, suggesting that this peptide motif can inhibit two subunits of Stx.
- Published
- 2021
14. Hepatitis B Virus Utilizes a Retrograde Trafficking Route via the Trans-Golgi Network to Avoid Lysosomal Degradation
- Author
-
Ying-Yi Li, Kazuyuki Kuroki, Tetsuro Shimakami, Kazuhisa Murai, Kazunori Kawaguchi, Takayoshi Shirasaki, Kouki Nio, Saiho Sugimoto, Tomoki Nishikawa, Hikari Okada, Noriaki Orita, Hideo Takayama, Ying Wang, Phuong Doan Thi Bich, Astuya Ishida, Sadahiro Iwabuchi, Shinichi Hashimoto, Takeshi Shimaoka, Noriko Tabata, Miho Watanabe-Takahashi, Kiyotaka Nishikawa, Hiroshi Yanagawa, Motoharu Seiki, Kouji Matsushima, Taro Yamashita, Shuichi Kaneko, and Masao Honda
- Subjects
Hepatology ,Gastroenterology - Abstract
Hepatitis B virus (HBV) infection is difficult to cure owing to the persistence of covalently closed circular viral DNA (cccDNA). We performed single-cell transcriptome analysis of newly established HBV-positive and HBV-negative hepatocellular carcinoma cell lines and found that dedicator of cytokinesis 11 (DOCK11) was crucially involved in HBV persistence. However, the roles of DOCK11 in the HBV lifecycle have not been clarified.The cccDNA levels were measured by Southern blotting and real-time detection polymerase chain reaction in various hepatocytes including PXB cells by using an HBV-infected model. The retrograde trafficking route of HBV capsid was investigated by super-resolution microscopy, proximity ligation assay, and time-lapse analysis. The downstream molecules of DOCK11 and underlying mechanism were examined by liquid chromatography-tandem mass spectrometry, immunoblotting, and enzyme-linked immunosorbent assay.The cccDNA levels were strongly increased by DOCK11 overexpression and repressed by DOCK11 suppression. Interestingly, DOCK11 functionally associated with retrograde trafficking proteins in the trans-Golgi network (TGN), Arf-GAP with GTPase domain, ankyrin repeat, and pleckstrin homology domain-containing protein 2, and ADP-ribosylation factor 1, together with HBV capsid, to open an alternative retrograde trafficking route for HBV from early endosomes (EEs) to the TGN and then to the endoplasmic reticulum (ER), thereby avoiding lysosomal degradation. Clinically, DOCK11 levels in liver biopsies from patients with chronic hepatitis B were significantly reduced by entecavir treatment, and this reduction correlated with HBV surface antigen levels.HBV uses a retrograde trafficking route via EEs-TGN-ER for infection that is facilitated by DOCK11 and serves to maintain cccDNA. Therefore, DOCK11 is a potential therapeutic target to prevent persistent HBV infection.
- Published
- 2021
15. Synthetic construction of sugar-amino acid hybrid polymers involving globotriaose or lactose and evaluation of their biological activities against Shiga toxins produced by Escherichia coli O157:H7
- Author
-
Miho Watanabe-Takahashi, Ken Hatano, Yusuke Goshu, Takahiko Matsushita, Yasuhiro Natori, Koji Matsuoka, Kiyotaka Nishikawa, Tetsuo Koyama, and Daiyo Terunuma
- Subjects
0301 basic medicine ,Polymers ,Clinical Biochemistry ,Radical polymerization ,Pharmaceutical Science ,Lactose ,Escherichia coli O157 ,Shiga Toxins ,010402 general chemistry ,medicine.disease_cause ,01 natural sciences ,Biochemistry ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Discovery ,medicine ,Copolymer ,Amino Acids ,Molecular Biology ,Escherichia coli ,chemistry.chemical_classification ,Dose-Response Relationship, Drug ,Molecular Structure ,Molecular mass ,Chemistry ,Organic Chemistry ,Amino Sugars ,Combinatorial chemistry ,0104 chemical sciences ,Amino acid ,030104 developmental biology ,Monomer ,Polymerization ,Molecular Medicine ,Radical initiator ,Trisaccharides - Abstract
Synthetic assembly of sugar moieties and amino acids in order to create “sugar-amino acid hybrid polymers” was accomplished by means of simple radical polymerization of carbohydrate monomers having an amino acid-modified polymerizable aglycon. Amines derived from globotriaoside and lactoside as glycoepitopes were condensed with known carbobenzyloxy derivatives, including Z-Gly, Z- l -Ala and Z-β-Ala, which had appropriate spacer ability and a chiral center to afford fully protected sugar-amino acid hybrid compounds in good yields. After deprotection followed by acryloylation, the water-soluble glycomonomers were polymerized with or without acrylamide in the presence of a radical initiator in water to give corresponding copolymers and homopolymers, which were shown by SEC analysis to have high molecular weights. Evaluation of the biological activities of the glycopolymers against Shiga toxins (Stxs) was carried out, and the results suggested that glycopolymers having highly clustered globotriaosyl residues had high affinity against Stx2 (KD = 2.7∼4.0 µM) even though other glycopolymers did not show any affinity or showed very weak binding affinity. When Stx1 was used for the same assay, all of the glycopolymers having globotriaosyl residues showed high affinity (KD = 0.30∼1.74 µM). Interestingly, couple of glycopolymers having lactosyl moieties had weaker binding affinity against Stx1. In addition, when cytotoxicity assays were carried out for both Stxs, glycopolymers having highly clustered globotriaosyl residues showed higher affinity than that of the copolymers, and only highly clustered-type glycopolymers displayed neutralization potency against Stx2.
- Published
- 2018
- Full Text
- View/download PDF
16. Acquired Resistance to Shiga Toxin-Induced Apoptosis by Loss of CD77 Expression in Human Myelogenous Leukemia Cell Line, THP-1
- Author
-
Takayuki Hattori, Miho Watanabe-Takahashi, Mikihiko Naito, and Kiyotaka Nishikawa
- Subjects
0301 basic medicine ,THP-1 Cells ,Pharmaceutical Science ,Apoptosis ,Biology ,Shiga Toxin 1 ,Shiga Toxin 2 ,Virulence factor ,03 medical and health sciences ,Myelogenous ,Downregulation and upregulation ,hemic and lymphatic diseases ,medicine ,Humans ,THP1 cell line ,Etoposide ,Pharmacology ,Trihexosylceramides ,Shiga toxin ,General Medicine ,Galactosyltransferases ,medicine.disease ,Antineoplastic Agents, Phytogenic ,Leukemia ,030104 developmental biology ,Leukemia, Myeloid ,Toxicity ,Cancer research ,biology.protein - Abstract
Shiga toxin (Stx) is a main virulence factor of Enterohemorrhagic Escherichia coli (EHEC) that causes diarrhea and hemorrhagic colitis and occasionally fatal systemic complications. Stx induces rapid apoptotic cell death in some cells, such as human myelogenous leukemia THP-1 cells expressing CD77, a receptor for Stx internalization, and the induction of apoptotic cell death is thought to be crucial for the fatal systemic complications. Therefore, in order to suppress the fatal toxicity, it is important to understand the mechanism how cells can escape from apoptotic cell death in the presence of Stx. In this study, we isolated resistant clones to Stx-induced apoptosis from highly sensitive THP-1 cells by continuous exposure with lethal dose of Stx. All of the ten resistant clones lost the expression of CD77 as a consequence of the reduction in CD77 synthase mRNA expression. These results suggest that downregulation of CD77 or CD77 synthase expression could be a novel approach to suppress the fatal toxicity of Stx in EHEC infected patient.
- Published
- 2018
- Full Text
- View/download PDF
17. Tocopherol suppresses 24(S)-hydroxycholesterol-induced cell death via inhibition of CaMKII phosphorylation
- Author
-
Yasuomi Urano, Minori Asa, Kiyotaka Nishikawa, Yoshiro Saito, Yuki Kimura, and Noriko Noguchi
- Subjects
0301 basic medicine ,Programmed cell death ,chemistry.chemical_element ,Tocopherols ,Calcium ,environment and public health ,Biochemistry ,03 medical and health sciences ,RIPK1 ,Ca2+/calmodulin-dependent protein kinase ,Cell Line, Tumor ,Humans ,Phosphorylation ,RNA, Small Interfering ,Protein kinase A ,Egtazic Acid ,Protein Kinase Inhibitors ,Neurons ,Gene knockdown ,Cell Death ,Chemistry ,General Medicine ,Hydroxycholesterols ,Cell biology ,Enzyme Activation ,enzymes and coenzymes (carbohydrates) ,030104 developmental biology ,Receptor-Interacting Protein Serine-Threonine Kinases ,sense organs ,Calcium-Calmodulin-Dependent Protein Kinase Type 2 ,Homeostasis ,Signal Transduction - Abstract
Although 24(S)-hydroxycholesterol (24S-OHC) plays an important role to maintain homeostasis of cholesterol in the brain, it induces neuronal cell death at high concentrations. 24S-OHC-induced cell death was suppressed by γ-tocopherol (γ-Toc) but not by γ-tocotrienol (γ-Toc3) in a similar way to α-tocopherol (α-Toc) and α-tocotrienol (α-Toc3) in human neuroblastoma SH-SY5Y cells. Both γ-Toc and γ-Toc3 significantly inhibited cumene hydroperoxide-induced cell death, as previously shown in the case of α-Toc and α-Toc3. Lipid droplet-like structure formation induced by 24S-OHC was suppressed by neither γ-Toc nor γ-Toc3. The phosphorylation of calcium/calmodulin-dependent protein kinase II (CaMKII) was induced by 24S-OHC, which was suppressed by CaMKII phosphorylation-site inhibitor mM3 but not by calmodulin-binding-site inhibitor KN62. A calcium chelator, BAPTA-AM, inhibited calcium ionophore A23187-induced CaMKII phosphorylation but not 24S-OHC-induced CaMKII phosphorylation. Receptor-interacting protein kinase 1 (RIPK1) phosphorylation induced by 24S-OHC was not inhibited by either mM3 or KN62, suggesting that CaMKII activation does not affect RIPK1 phosphorylation. Knockdown of RIPK1 using siRNA induced not only inhibition of CaMKII phosphorylation but also reduction of total CaMKII protein levels, suggesting that RIPK1 may regulate CaMKII signalling. 24S-OHC-induced RIPK1 phosphorylation was inhibited by neither α-Toc nor α-Toc3. In contrast, CaMKII phosphorylation induced by 24S-OHC was significantly suppressed by α-Toc but not by α-Toc3. These results suggest that CaMKII activation is involved in the mechanism of 24S-OHC-induced cell death and that Toc inhibits the cell death via inhibition of CaMKII activation through a RIPK1 phosphorylation-independent pathway.
- Published
- 2018
18. Proteasome inhibitors prevent cell death and prolong survival of mice challenged by Shiga toxin
- Author
-
Kiyotaka Nishikawa, Takayuki Hattori, Nobumichi Ohoka, Mikihiko Naito, Takashi Hamabata, Miho Watanabe-Takahashi, and Koichi Furukawa
- Subjects
Programmed cell death ,Mcl-1, myeloid cell leukemia 1 ,QH301-705.5 ,Poly ADP ribose polymerase ,Apoptosis inhibitory proteins ,Apoptosis ,Biology ,General Biochemistry, Genetics and Molecular Biology ,ER, endoplasmic reticulum ,PI, propidium iodide ,CHX, cycloheximide ,hemic and lymphatic diseases ,Research article ,medicine ,Proteasome inhibitor ,Biology (General) ,STEC, Shiga toxin-producing Escherichia coli ,c-IAP1, cellular inhibitor of apoptosis protein 1 ,FLIP, FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein ,Caspase-9 ,Proteasome ,Bortezomib ,Shiga toxin ,BRZ, bortezomib ,Immunology ,biology.protein ,Cancer research ,Stx, Shiga toxin ,PARP, Poly(ADP-ribose) polymerase ,medicine.drug - Abstract
Highlights • Shiga toxin (Stx) rapidly reduces the level of short-lived anti-apoptotic proteins. • Stx induces activation of caspase 9 and apoptosis. • Proteasome inhibitors prevent the reduction of anti-apoptotic proteins. • Proteasome inhibitors suppress Stx-induced apoptosis. • Bortezomib prolongs the survival of mice challenged with a lethal dose of Stx., 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.
- Published
- 2015
19. Pleckstrin homology domain of p210 BCR-ABL interacts with cardiolipin to regulate its mitochondrial translocation and subsequent mitophagy
- Author
-
Kiyotaka Nishikawa, Kentaro Shimasaki, Keigo Kumagai, Yoshiro Saito, Masato Umeda, Norihito Shibata, Kentaro Hanada, Satoru Funamoto, Noriko Noguchi, Yoshiro Maru, Miho Watanabe-Takahashi, Mikihiko Naito, and Fujiko Tsukahara
- Subjects
0301 basic medicine ,Carbonyl Cyanide m-Chlorophenyl Hydrazone ,Cardiolipins ,Fusion Proteins, bcr-abl ,Biology ,Mitochondrion ,Philadelphia chromosome ,03 medical and health sciences ,chemistry.chemical_compound ,Cytosol ,hemic and lymphatic diseases ,Mitophagy ,Genetics ,medicine ,Cardiolipin ,Humans ,Pleckstrin Homology Domains ,Cell Biology ,medicine.disease ,Fusion protein ,Cell biology ,Mitochondria ,Pleckstrin homology domain ,Protein Transport ,030104 developmental biology ,HEK293 Cells ,chemistry ,Protein kinase domain - Abstract
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 organelle's 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 domain's contribution to CML pathogenesis.
- Published
- 2017
20. Exosome-associated Shiga toxin 2 is released from cells and causes severe toxicity in mice
- Author
-
Hirofumi Ukai, Noriko Toyama-Sorimachi, Shinji Yamasaki, Masayuki Murata, Kiyotaka Nishikawa, Waka Sato, Kentaro Shimasaki, Yasuhiro Natori, Jumpei Omi, Yuri Nishino, Atsuo Miyazawa, Miho Watanabe-Takahashi, Jyoji Yamate, Fumi Kano, Miwa Tamura-Nakano, Ryohei Yanoshita, Jun Motoyama, and Masaya Ikegawa
- Subjects
0301 basic medicine ,Endosome ,Virulence Factors ,lcsh:Medicine ,Endosomes ,medicine.disease_cause ,Exosomes ,Kidney ,Exosome ,Shiga Toxin 2 ,Virulence factor ,Article ,Microbiology ,03 medical and health sciences ,Mice ,fluids and secretions ,In vivo ,hemic and lymphatic diseases ,medicine ,Animals ,lcsh:Science ,Escherichia coli ,Multidisciplinary ,biology ,Chemistry ,lcsh:R ,Shiga toxin ,Biological Transport ,biochemical phenomena, metabolism, and nutrition ,bacterial infections and mycoses ,Microvesicles ,Epithelium ,030104 developmental biology ,medicine.anatomical_structure ,biology.protein ,bacteria ,lcsh:Q - Abstract
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.
- Published
- 2017
21. Affinity-Based Screening of Tetravalent Peptides Identifies Subtype-Selective Neutralizers of Shiga Toxin 2d, a Highly Virulent Subtype, by Targeting a Unique Amino Acid Involved in Its Receptor Recognition
- Author
-
Shinji Yamasaki, Kiyotaka Nishikawa, Satoru Funamoto, Miho Watanabe-Takahashi, Baihao Zhang, Eiko Shimizu, and Mitsui Takaaki
- Subjects
0301 basic medicine ,Pentamer ,Virulence Factors ,030106 microbiology ,Immunology ,Virulence ,Peptide ,medicine.disease_cause ,Microbiology ,Shiga Toxin 2 ,Virulence factor ,Cell Line ,03 medical and health sciences ,Mice ,Peptide Library ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Amino Acids ,Peptide library ,Escherichia coli ,Vero Cells ,Escherichia coli Infections ,chemistry.chemical_classification ,Cellular Microbiology: Pathogen-Host Cell Molecular Interactions ,biology ,Shiga toxin ,Molecular biology ,Amino acid ,Mice, Inbred C57BL ,030104 developmental biology ,Infectious Diseases ,chemistry ,Biochemistry ,Enterohemorrhagic Escherichia coli ,biology.protein ,Parasitology ,Peptides ,Protein Binding - Abstract
Shiga toxin (Stx), a major virulence factor of enterohemorrhagic Escherichia coli (EHEC), can be classified into two subgroups, Stx1 and Stx2, each consisting of various closely related subtypes. Stx2 subtypes Stx2a and Stx2d are highly virulent and linked with serious human disorders, such as acute encephalopathy and hemolytic-uremic syndrome. Through affinity-based screening of a tetravalent peptide library, we previously developed peptide neutralizers of Stx2a in which the structure was optimized to bind to the B-subunit pentamer. In this study, we identified Stx2d-selective neutralizers by targeting Asn16 of the B subunit, an amino acid unique to Stx2d that plays an essential role in receptor binding. We synthesized a series of tetravalent peptides on a cellulose membrane in which the core structure was exactly the same as that of peptides in the tetravalent library. A total of nine candidate motifs were selected to synthesize tetravalent forms of the peptides by screening two series of the tetravalent peptides. Five of the tetravalent peptides effectively inhibited the cytotoxicity of Stx2a and Stx2d, and notably, two of the peptides selectively inhibited Stx2d. These two tetravalent peptides bound to the Stx2d B subunit with high affinity dependent on Asn16. The mechanism of binding to the Stx2d B subunit differed from that of binding to Stx2a in that the peptides covered a relatively wide region of the receptor-binding surface. Thus, this highly optimized screening technique enables the development of subtype-selective neutralizers, which may lead to more sophisticated treatments of infections by Stx-producing EHEC.
- Published
- 2016
22. M-COPA, a novel Golgi system disruptor, suppresses apoptosis induced by Shiga toxin
- Author
-
Shingo Dan, Isamu Shiina, Yoshimi Ohashi, Takao Yamori, Takayuki Hattori, Kiyotaka Nishikawa, Miho Watanabe-Takahashi, and Mikihiko Naito
- Subjects
0301 basic medicine ,Diarrhea ,Pyridines ,media_common.quotation_subject ,Antidotes ,Golgi Apparatus ,Apoptosis ,Naphthols ,Alkenes ,medicine.disease_cause ,Virulence factor ,Shiga Toxin ,03 medical and health sciences ,symbols.namesake ,hemic and lymphatic diseases ,Genetics ,medicine ,Humans ,Internalization ,Escherichia coli ,Caspase ,media_common ,biology ,Shiga-Toxigenic Escherichia coli ,Shiga toxin ,Cell Biology ,Golgi apparatus ,Colitis ,Cell biology ,Vesicular transport protein ,030104 developmental biology ,biology.protein ,symbols ,ADP-Ribosylation Factor 1 - Abstract
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.
- Published
- 2016
23. Rescue from lethal Shiga toxin 2-induced renal failure with a cell-permeable peptide
- Author
-
Diann Debord, Caitlin S. Leibowitz, Scott Freeman, Kiyotaka Nishikawa, Shinichiro Kurosawa, Valta Collins, Deborah J. Stearns-Kurosawa, and Sun-Young Oh
- Subjects
Male ,Nephrology ,medicine.medical_specialty ,Anemia ,medicine.drug_class ,Antibiotics ,Fluorescent Antibody Technique ,Pharmacology ,medicine.disease_cause ,Shiga Toxin 2 ,Article ,chemistry.chemical_compound ,Internal medicine ,medicine ,Animals ,Escherichia coli Infections ,Creatinine ,biology ,business.industry ,Toxin ,Acute kidney injury ,Shiga toxin ,Acute Kidney Injury ,medicine.disease ,Disease Models, Animal ,chemistry ,Concomitant ,Hemolytic-Uremic Syndrome ,Pediatrics, Perinatology and Child Health ,Immunology ,biology.protein ,Cytokines ,Peptides ,business ,Papio - Abstract
Intestinal infection with Shiga toxin (Stx)-producing E.coli is a leading cause of hemolytic uremic syndrome and acute renal injury in otherwise healthy children in the US. Antibiotics are contraindicated and a therapeutic priority is agents that act intracellularly against the bacterial toxins that drive kidney injury. Our aim was to evaluate whether intravenous administration of a cell-permeable peptide (TVP) that binds to Stx2 will reduce disease severity and rescue juvenile baboons from a lethal Stx2 dose (50 ng/kg). TVP (5 mg/kg) was delivered i.v. simultaneously with toxin (prevention protocol) or at 6 or 24 h after toxin with daily 1 mg/kg supplements up to day 4 (rescue protocols). Biomarkers were monitored in blood and urine up to 28 days. TVP therapy resulted in either absence of clinical signs of acute kidney injury and normal urine output (prevention), or delayed and reduced BUN and creatinine levels (rescue) with concomitant survival. Delayed peptide administration significantly reduced thrombocytopenia, but surprisingly did not alter anemia even when monitored for 28 days in rescued survivors. This is the first successful cell-permeable therapeutic that counteracts Stx2 lethality in an animal model, which recapitulates many of the human responses to enteric infection.
- Published
- 2011
- Full Text
- View/download PDF
24. An Orally Applicable Shiga Toxin Neutralizer Functions in the Intestine To Inhibit the Intracellular Transport of the Toxin
- Author
-
Kiyotaka Nishikawa, Taeko Dohi, Miho Watanabe-Takahashi, Yasuhiro Natori, Fumi Kano, Takashi Hamabata, Masayuki Murata, Toshio Sato, and Noriko Noguchi
- Subjects
animal diseases ,Immunology ,Administration, Oral ,Ileum ,Biology ,medicine.disease_cause ,Shiga Toxin 2 ,Microbiology ,Virulence factor ,Tissue Culture Techniques ,symbols.namesake ,chemistry.chemical_compound ,fluids and secretions ,Shiga-like toxin ,medicine ,Animals ,Humans ,Escherichia coli ,Endoplasmic reticulum ,Biological Transport ,Shiga toxin ,biochemical phenomena, metabolism, and nutrition ,Golgi apparatus ,bacterial infections and mycoses ,Infectious Diseases ,medicine.anatomical_structure ,chemistry ,Caco-2 ,Microbial Immunity and Vaccines ,symbols ,biology.protein ,bacteria ,Parasitology ,Rabbits ,Caco-2 Cells ,Peptides - 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.
- Published
- 2010
- Full Text
- View/download PDF
25. Yip1A regulates the COPI-independent retrograde transport from the Golgi complex to the ER
- Author
-
Masayuki Murata, Fumi Kano, Kiyotaka Nishikawa, Shinobu Yamauchi, Miho Watanabe-Takahashi, Yumi Yoshida, and Nobuhiro Nakamura
- Subjects
Recombinant Fusion Proteins ,Vesicular Transport Proteins ,Golgi Apparatus ,Biology ,Endoplasmic Reticulum ,Shiga Toxin 1 ,Cell Line ,Coat Protein Complex I ,symbols.namesake ,Viral Envelope Proteins ,RNA interference ,Animals ,Humans ,Galactosyltransferase ,Gene knockdown ,Membrane Glycoproteins ,Endoplasmic reticulum ,Biological Transport ,Cell Biology ,COPI ,Golgi apparatus ,Cell biology ,Membrane protein ,rab GTP-Binding Proteins ,symbols ,Axoplasmic transport ,RNA Interference ,Peptides ,HeLa Cells - Abstract
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.
- Published
- 2009
- Full Text
- View/download PDF
26. Type I platelet-activating factor acetylhydrolase catalytic subunits over-expression induces pleiomorphic nuclei and centrosome amplification
- Author
-
Hiroyuki Koizumi, Noritaka Yamaguchi, Hiroyuki Arai, Junken Aoki, Yasuhiro Natori, Kiyotaka Nishikawa, Yumiko Natori, and Yasukazu Takanezawa
- Subjects
Dynein ,CHO Cells ,Biology ,Type I lissencephaly ,Models, Biological ,Catalysis ,Mice ,Cricetulus ,Microtubule ,Catalytic Domain ,Cricetinae ,Genetics ,Animals ,Cell Nucleus ,Centrosome ,Brain ,Cell Biology ,Phenotype ,Recombinant Proteins ,Cell biology ,Gene Expression Regulation ,Biochemistry ,1-Alkyl-2-acetylglycerophosphocholine Esterase ,Over expression ,Drosophila ,Microtubule-Associated Proteins ,Function (biology) - Abstract
LIS1, a causative gene product for type I lissencephaly, binds to and regulates the dynein motor and the centrosome. LIS1 also forms a complex with the catalytic subunits alpha1 and alpha2 of type I platelet-activating factor acetylhydrolase [PAF-AH (I)]. However, the cellular function of the catalytic subunits remains unknown. In this study, we showed that over-expression of the catalytic subunits, especially alpha2, in cultured cells induced dramatic phenotypical changes including nuclear shape change, centrosomal amplification and microtubule disorganization. We examined if these effects were due to the catalytic activity and/or binding of alpha2 to LIS1. Substitution of a single amino acid Glu39 of murine alpha1 and alpha2 by Asp (alpha2-E39D) did not affect catalytic activity but completely abolished LIS1 binding. Over-expression of either alpha2-E39D or the catalytically inactive alpha2-S48C revealed that alpha2-E39D, but not alpha2-S48C, lost its ability to induce above-mentioned phenotypic changes. Biochemical analyses showed that LIS1 present in the precipitate fraction of murine brain homogenates could be translocated to the soluble fraction by alpha2, but not by alpha2-E39D. These results suggest that over-expression of the PAF-AH (I) catalytic subunits induces centrosomal amplification and microtubule disorganization by disturbing intracellular localization of LIS1.
- Published
- 2007
- Full Text
- View/download PDF
27. Development of a novel tetravalent synthetic peptide that binds to phosphatidic acid
- Author
-
Kiyotaka Nishikawa, Kohjiro Nagao, Kentaro Taniuchi, Utako Kato, Takehiko Inaba, Kazunari Akiyoshi, Yoshihiro Sasaki, Toshihide Kobayashi, Miho Watanabe-Takahashi, Yuji Hara, Rina Ogawa, Masato Umeda, and Masaki Tsuchiya
- Subjects
Stereochemistry ,Static Electricity ,lcsh:Medicine ,Phosphatidic Acids ,Peptide ,chemistry.chemical_compound ,Peptide Library ,Phosphatidylcholine ,lcsh:Science ,Peptide library ,Phosphatidylethanolamine ,chemistry.chemical_classification ,Membranes ,Multidisciplinary ,lcsh:R ,Tryptophan ,Hydrogen Bonding ,Phosphatidylserine ,Phosphatidic acid ,Amino acid ,Dissociation constant ,Kinetics ,Amino Acid Substitution ,chemistry ,Biochemistry ,lcsh:Q ,Peptides ,Research Article - Abstract
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 PABTP 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.
- Published
- 2015
28. Identification of a Wide Range of Motifs Inhibitory to Shiga Toxin by Affinity-Driven Screening of Customized Divalent Peptides Synthesized on a Membrane
- Author
-
Miho Watanabe-Takahashi, Kato Mihoko, Eiko Shimizu, and Kiyotaka Nishikawa
- Subjects
Pentamer ,Cell Survival ,Protein subunit ,Antidotes ,Plasma protein binding ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Divalent ,Shiga Toxin ,Chlorocebus aethiops ,medicine ,Methods ,Animals ,Peptide library ,Escherichia coli ,Vero Cells ,chemistry.chemical_classification ,Ecology ,biology ,Shiga toxin ,Amino acid ,Biochemistry ,chemistry ,biology.protein ,Peptides ,Food Science ,Biotechnology ,Protein Binding - 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.
- Published
- 2015
29. Development of dialyzer with immobilized glycoconjugate polymers for removal of Shiga-toxin
- Author
-
Kenichi Hatanaka, Atsushi Miyagawa, Katsura Igai, Maria Carmelita Z. Kasuya, Kiyotaka Nishikawa, Yasuhiro Natori, and Miho Watanabe
- Subjects
Magnetic Resonance Spectroscopy ,Time Factors ,Materials science ,Polymers ,Glycoconjugate ,Blotting, Western ,Carbohydrates ,Biophysics ,Biocompatible Materials ,Bioengineering ,Ligands ,Shiga Toxin ,Biomaterials ,chemistry.chemical_compound ,Adsorption ,Dialysis Solutions ,Chlorocebus aethiops ,Materials Testing ,Animals ,Fiber ,Cellulose ,Vero Cells ,chemistry.chemical_classification ,Chromatography ,Dose-Response Relationship, Drug ,technology, industry, and agriculture ,Membranes, Artificial ,Polymer ,Condensation reaction ,Hemodialysis Solutions ,Kinetics ,Membrane ,Models, Chemical ,chemistry ,Mechanics of Materials ,Covalent bond ,Ceramics and Composites ,Dialysis ,Glycoconjugates - Abstract
The dialyzer for Shiga-toxin elimination was developed and its performance was established. The dialyzer was prepared by immobilization of multivalent ligands. Glycoconjugate polymers having oligosaccharides and amino groups were synthesized to function as Shiga-toxin adsorbents. The amino group was utilized to immobilize the polymer inside the cellulose hollow fiber of the dialyzer. Cellulose hollow fibers packed in the dialyzer were carboxymethylated under moderate conditions. The glycoconjugate polymers were bound covalently to the hollow fibers of the dialyzer by condensation reaction between the amino group of the polymer and the carboxyl group of the cellulose hollow fiber. Shiga-toxin eliminabilities of the prepared dialyzers were evaluated at various conditions. Even at high concentration of protein such as FCS, the dialyzer showed an excellent performance for Shiga-toxin adsorption.
- Published
- 2006
- Full Text
- View/download PDF
30. Syntheses and Vero toxin-binding activities of carbosilane dendrimers periphery-functionalized with galabiose
- Author
-
Ken Hatano, Kiyotaka Nishikawa, Kumiko Hino, Yasuaki Esumi, Yasuhiro Natori, Daiyo Terunuma, Akihiro Yamada, Tetsuo Koyama, Koji Matsuoka, and Hiroyuki Koshino
- Subjects
Stereochemistry ,Chemistry ,Dendrimer ,Organic Chemistry ,Drug Discovery ,Kinetic analysis ,Vero Toxin ,medicine ,medicine.disease_cause ,Biochemistry ,Escherichia coli - Abstract
Carbosilane dendrimers bearing galabiose (Galα1–4Gal) with three, four, and six galabiose units at the periphery of the dendrimers were synthesized for use as artificial inhibitors against Shiga toxins (Stxs) produced by Escherichia coli O157:H7. The galabiose unit, prepared from penta-O-acetyl-β- d -galactopyranose, was linked with carbosilane dendrimers of three shapes to afford acetyl-protected glycodendrimers in good yields. De-O-acetylation of the clusters was carried out in the presence of NaOMe and then aq NaOH to give the desired three shapes of galabiose-coated carbosilane dendrimers. Their biological activities toward Stxs were evaluated by kinetic analysis, binding assays, and cytotoxic assays.
- Published
- 2006
- Full Text
- View/download PDF
31. Histidine-Tagged Shiga Toxin B Subunit Binding Assay : Simple and Specific Determination of Gb3 Content in Mammalian Cells
- Author
-
Satoshi Ishii, Hiroki Maruyama, Kiyotaka Nishikawa, and In-Sun Shin
- Subjects
Time Factors ,Cells ,Globotriaosylceramide ,binding assay method ,His-tagged protein ,medicine.disease_cause ,Glycosphingolipids ,law.invention ,HeLa ,Mice ,chemistry.chemical_compound ,Glycolipid ,law ,Drug Discovery ,medicine ,Animals ,Humans ,Histidine ,Peroxidase ,globotriaosylceramide ,Ganglioside ,biology ,Benzidines ,Trihexosylceramides ,Ligand binding assay ,Cholera toxin ,Heart ,Shiga toxin ,General Chemistry ,General Medicine ,Fibroblasts ,biology.organism_classification ,Molecular biology ,Biochemistry ,chemistry ,Recombinant DNA ,biology.protein ,HeLa Cells - Abstract
application/pdf, A two-step binding assay for globotriaosylceramide (Gb3) content was developed by histidine-tagging strategy, which is a well-established method for the purification of recombinant proteins. The complete binding of the recombinant His-tagged Shiga toxin 1B subunit (1B-His) (1 microg/ml) to the standard Gb3 adsorbed on a multi-well H type plate was observed within 30 min at 37 degrees C; and its binding could be visualized by the following applications of HisProbe-HRP (8 microg/ml) and tetramethylbenzidine (TMB) peroxidase substrate. The 1B-His binding assay was linear over the range of 1 to 100 ng of Gb3 per well. The binding of 1B-His was specific to Gb3 separated from HeLa cells, and no major cross-reactivity of other glycolipids in Folch's lower fractions extracted from HeLa cells was detected. The glycolipids in Folch's lower fractions from HeLa cells, human fibroblasts and mouse heart were suitable for this assay, but the further purification was needed for glycolipids from human plasma, thus sample preparation is critical factor for the reliable determination of Gb3 content. The 1B-His binding to Gb3 was inhibited by the addition of galactose, but not mannose. This 1B-His binding assay will be useful not only for the determination of Gb3 content, but also for screening for the compounds which inhibit the toxin-binding to Gb3. The strategy of our present method may be applicable for other binding assay, such as Cholera toxin B-subunit for ganglioside GM1., 日本薬学会 Chemical & pharmaceutical bulletin 54(4) p.522-527(2006)より転載
- Published
- 2006
32. Structural Analysis of the Interaction between Shiga Toxin B Subunits and Linear Polymers Bearing Clustered Globotriose Residues
- Author
-
Yasuhiro Natori, Miho Watanabe, Koji Matsuoka, Katsura Igai, Daiyo Terunuma, Yuji Samejima, Toshiyuki Watanabe, Ryohei Yanoshita, Atsushi Miyagawa, and Kiyotaka Nishikawa
- Subjects
Polymers ,Protein subunit ,Immunology ,Globotriaosylceramide ,Escherichia coli O157 ,Shiga Toxin 1 ,Shiga Toxin 2 ,Microbiology ,chemistry.chemical_compound ,fluids and secretions ,Shiga-like toxin ,STX2 ,Chlorocebus aethiops ,Animals ,Trisaccharide ,Binding site ,Vero Cells ,chemistry.chemical_classification ,Acrylamide ,biology ,Shiga toxin ,Polymer ,Molecular Pathogenesis ,Protein Subunits ,Infectious Diseases ,chemistry ,Biochemistry ,biology.protein ,Parasitology ,Trisaccharides - Abstract
We previously developed linear polymers bearing clustered trisaccharides of globotriaosylceramide (Gb3) as orally applicable Shiga toxin (Stx) neutralizers. Here, using a Gb3 polymer with a short spacer tethering the trisaccharide to the core, we found that shortening the spacer length markedly reduced the binding affinity for Stx2 but not Stx1. Moreover, mutational analysis revealed that the essential binding sites of the terminal trisaccharides were completely different between Stx1 and Stx2. These results provide the molecular basis for the interaction between Stx B subunits and Gb3 polymers.
- Published
- 2006
- Full Text
- View/download PDF
33. Shiga Toxin 1 Causes Direct Renal Injury in Rats
- Author
-
Kiyotaka Nishikawa, Shinobu Miyazawa, Seiichi Matsuo, Yasuhiro Natori, Elise T. Yamamoto, Lianshan Zhang, and Masashi Mizuno
- Subjects
Male ,medicine.medical_specialty ,Platelet Aggregation ,Immunology ,Gene Expression ,Renal function ,Apoptosis ,Medullary interstitium ,Biology ,Kidney ,Shiga Toxin 1 ,Microbiology ,Blood Urea Nitrogen ,chemistry.chemical_compound ,Internal medicine ,medicine.artery ,medicine ,Animals ,RNA, Messenger ,Rats, Wistar ,Renal artery ,Blood urea nitrogen ,Creatinine ,Tumor Necrosis Factor-alpha ,urogenital system ,Molecular Pathogenesis ,Rats ,Infectious Diseases ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Kidney Diseases ,Parasitology ,Renal vein ,Immunostaining - Abstract
Infection with Shiga toxin (Stx)-producing Escherichia coli has been implicated to cause hemolytic uremic syndrome, which is characterized by histological abnormalities such as microvascular thrombi and tubular cell damage in the kidney. Although Stx is known to be the major virulence factor of the pathogen, it is still unclear whether Stx directly impairs renal cells in vivo to cause such histological changes and deterioration of renal function. To assess the consequence of the direct action of Stx on renal cells, left kidneys of rats were perfused with Stx1 from the renal artery through the renal vein and then revascularized. Kidneys of control animals were perfused with the vehicle alone. On day 1, apoptosis and induction of tumor necrosis factor alpha gene expression were noticed to occur in the medulla of the Stx1-perfused kidneys. On day 3, extensive tubular injuries were observed by light microscopy: aggregated platelets and monocytic infiltrates in both glomeruli and the medullary interstitium were detected by immunostaining. Tubular changes were more extensive on day 9, with areas of infarction seen in the cortex and medulla. These changes were not found to occur in the sham-operated kidneys. No obvious glomerular changes were detected by light microscopy at any time point. When nonperfused right kidneys were removed after the Stx1 perfusion of the left kidneys, the serum creatinine and blood urea nitrogen levels were increased from day 2, and acute renal failure followed on day 3. These results indicate that Stx1 caused glomerular platelet aggregation, tubular damage, and acute deterioration of renal function by acting directly on renal cells.
- Published
- 2005
- Full Text
- View/download PDF
34. Identification of the Optimal Structure Required for a Shiga Toxin Neutralizer with Oriented Carbohydrates to Function in the Circulation
- Author
-
Kumiko Hino, Daiyo Terunuma, Miho Watanabe, Yasuhiro Natori, Kiyotaka Nishikawa, Akihiro Yamada, Katsura Igai, Nobuhisa Abe, Hiroyoshi Kuzuhara, Ken Hatano, and Koji Matsuoka
- Subjects
Time Factors ,Structural similarity ,Carbohydrates ,medicine.disease_cause ,Virulence factor ,Shiga Toxin ,Twig ,Mice ,chemistry.chemical_compound ,Shiga-like toxin ,hemic and lymphatic diseases ,Chlorocebus aethiops ,parasitic diseases ,Carbohydrate Conformation ,Escherichia coli ,medicine ,Animals ,Immunology and Allergy ,natural sciences ,Binding site ,Vero Cells ,Mice, Inbred ICR ,biology ,Shiga toxin ,Kinetics ,Protein Subunits ,Infectious Diseases ,chemistry ,Biochemistry ,Drug Design ,biology.protein ,Female ,Function (biology) ,Protein Binding - Abstract
Shiga toxin (Stx) is a major virulence factor of Stx-producing Escherichia coli. Recently, we developed a therapeutic Stx neutralizer with 6 trisaccharides of globotriaosyl ceramide, a receptor for Stx, in its dendrimer structure (referred to as "SUPER TWIG [1]6") to function in the circulation. Here, we determined the optimal structure of SUPER TWIG for it to function in the circulation and identified a SUPER TWIG with 18 trisaccharides, SUPER TWIG (2)18, as another potent Stx neutralizer. SUPER TWIGs (1)6 and (2)18 shared a structural similarity, a dumbbell shape in which 2 clusters of trisaccharides were connected via a linkage with a hydrophobic chain. The dumbbell shape was found to be required for formation of a complex with Stx that enables efficient uptake and degradation of Stx by macrophages and, consequently, for potent Stx-neutralizing activity in the circulation. We also determined the binding site of the SUPER TWIGs on Stx.
- Published
- 2005
- Full Text
- View/download PDF
35. Induction of cytokines by toxins that have an identical RNA N-glycosidase activity: Shiga toxin, ricin, and modeccin
- Author
-
Tatsuya Oda, Yumiko Natori, Yukie Katayama, Nobukazu Komatsu, Yasuhiro Natori, Kiyotaka Nishikawa, Chisato Yamasaki, and Xun-Ting Zeng
- Subjects
Ribosome Inactivating Proteins ,Biophysics ,Lactose ,Ricin ,Cycloheximide ,Biochemistry ,Shiga Toxin ,chemistry.chemical_compound ,Chlorocebus aethiops ,Protein biosynthesis ,Animals ,Humans ,N-Glycosyl Hydrolases ,Vero Cells ,Molecular Biology ,Protein Synthesis Inhibitors ,Diphtheria toxin ,Sphingolipids ,Brefeldin A ,biology ,Ribosome-inactivating protein ,Interleukin-8 ,RNA ,Shiga toxin ,Molecular biology ,Ribosome Inactivating Proteins, Type 2 ,Protein Subunits ,chemistry ,Liposomes ,biology.protein ,Cytokines ,Caco-2 Cells ,Glycolipids ,Plant Lectins - Abstract
Shiga toxin (Stx) has an A1-B5 subunit structure, and the A subunit is an RNA N-glycosidase that inhibits cellular protein synthesis. We previously reported that in Caco-2 cells Stx induced cytokines and that the RNA N-glycosidase activity was essential for the cytokine induction. It is known that the binding of the Stx-B subunit to its receptor glycolipid, Gb3, mediates an A subunit-independent signal in some types of cells, but the involvement of this signal in the cytokine induction is unclear. In this study, we investigated whether RNA N-glycosidase itself induces cytokines. IL-8 production was enhanced by Stx, ricin, and modeccin, three toxins that inhibit protein synthesis through an identical RNA N-glycosidase activity, but not by two other types of protein synthesis inhibitors, diphtheria toxin and cycloheximide. The RNA N-glycosidase-type toxins showed a similar induction pattern of cytokine mRNAs. Brefeldin A, a Golgi apparatus inhibitor, completely suppressed the cytokine induction by the toxins. Analysis by using inhibitors of toxin binding and also Stx-B subunit showed that the cytokine-inducing activity was independent of Gb3-mediated signaling. These results indicate that RNA N-glycosidase itself induces the cytokine production and that intracellular transport of toxins through the Golgi apparatus is essential for the activity.
- Published
- 2004
- Full Text
- View/download PDF
36. A therapeutic agent with oriented carbohydrates for treatment of infections by Shiga toxin-producing Escherichia coli O157:H7
- Author
-
Masahiro Nishijima, Chisato Yamasaki, Kumiko Hino, Daiyo Terunuma, Hiroyoshi Kuzuhara, Yasuhiro Natori, Kiyotaka Nishikawa, Noriko Okabe, Junken Aoki, Eiji Kita, Yoshio Yamakawa, Koji Matsuoka, Masashi Mizuguchi, Sachio Takashima, and Shinobu Miyazawa
- Subjects
Cell Survival ,Recombinant Fusion Proteins ,Carbohydrates ,Escherichia coli O157 ,Transfection ,medicine.disease_cause ,Shiga Toxin ,Twig ,Microbiology ,Mice ,In vivo ,hemic and lymphatic diseases ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Vero Cells ,Escherichia coli ,Escherichia coli Infections ,Glutathione Transferase ,Multidisciplinary ,biology ,Toxin ,Shiga toxin ,Mononuclear phagocyte system ,Biological Sciences ,Silanes ,Virology ,Anti-Bacterial Agents ,Drug Design ,Macrophages, Peritoneal ,biology.protein ,Vero cell ,Trisaccharides - Abstract
Infection with Shiga toxin (Stx)-producing Escherichia coli O157:H7, which causes diarrhea and hemorrhagic colitis in humans, often results in fatal systemic complications, such as neurological damage and hemolytic–uremic syndrome. Because Stx circulating in the blood is a major causative factor of these complications, the development of a Stx neutralizer that functions in the circulation holds promise as a viable therapy. Here we developed a series of carbosilane dendrimers, in which trisaccharides of globotriaosyl ceramide, a receptor for Stx, were variously oriented at their termini (referred to as SUPER TWIG), and identified a SUPER TWIG with six trisaccharides as a Stx neutralizer functioning in the circulation. This SUPER TWIG specifically bound to Stx with high affinity ( K d = 1.1 × 10 −6 M) and inhibited the incorporation of the toxin into target cells. Intravenous administration of the SUPER TWIG along with Stx to mice substantially reduced the fatal brain damage and completely suppressed the lethal effect of Stx. Moreover, the SUPER TWIG protected mice from challenge with a fatal dose of E. coli O157:H7, even when administered after the establishment of the infection. The SUPER TWIG neutralized Stx in vivo by a mechanism in which the accumulation and immediate degradation of Stx by phagocytic macrophages present in the reticuloendothelial system were induced. Taken together, our findings indicate that this SUPER TWIG is therapeutic agent against infections by Stx-producing E. coli .
- Published
- 2002
- Full Text
- View/download PDF
37. A Peptide Library Approach Identifies a Specific Inhibitor for the ZAP-70 Protein Tyrosine Kinase
- Author
-
Michael B. Yaffe, Kiyotaka Nishikawa, Jack Lai, Steven J. Burakoff, Sansana Sawasdikosol, David A. Fruman, Zhou Songyang, and Lewis C. Cantley
- Subjects
T-Lymphocytes ,Molecular Sequence Data ,Syk ,chemical and pharmacologic phenomena ,Peptide ,Biology ,Lymphocyte Activation ,Transfection ,Jurkat Cells ,Genes, Reporter ,Peptide Library ,Humans ,Amino Acid Sequence ,Enzyme Inhibitors ,Kinase activity ,Protein kinase A ,Peptide library ,Molecular Biology ,chemistry.chemical_classification ,ZAP-70 Protein-Tyrosine Kinase ,Phospholipase C gamma ,Kinase ,hemic and immune systems ,Cell Biology ,Protein-Tyrosine Kinases ,Molecular biology ,Amino acid ,Isoenzymes ,Kinetics ,chemistry ,Biochemistry ,Type C Phospholipases ,Interleukin-2 ,Phosphorylation ,Peptides - Abstract
Summary cally blocked signaling downstream of ZAP-70. Our studies extend the range of experimental approaches We utilized a novel peptide library approach to identify specific inhibitors of ZAP-70, a protein Tyr kinase in- for probing ZAP-70 function in vivo and introduce a technique for developing protein kinase inhibitors that volved in T cell activation. By screening more than 6 billion peptides oriented by a common Tyr residue could be useful for a wide range of protein kinases. for their ability to bind to ZAP-70, we determined a consensus optimal peptide. A Phe-for-Tyr substituted Results and Discussion version of the peptide inhibited ZAP-70 protein Tyr kinase activity by competing with protein substrates In order to identify high-affinity inhibitors of the ZAP-70 (KI of 2 mM). The related protein Tyr kinases, Lck and protein Tyr kinase, we screened a Tyr-oriented peptide Syk, were not significantly inhibited by the peptide. library by affinity purification rather than by catalytic When introduced into intact T cells, the peptide conversion. The peptide library used contained the seblocked signaling downstream of ZAP-70, including quence Met-Ala-X-X-X-X-Tyr-X-X-X-X-Ala-Lys-Lys-Lys ZAP-70-dependent gene induction, without affecting where X indicates all amino acids except Trp, Cys, or upstream Tyr phosphorylation. Thus, screening Tyr- Tyr. The predicted degeneracy of this library is 178 < oriented peptide libraries can identify selective pep- 6.9 billion. Screening was performed in the presence of tide inhibitors of protein Tyr kinases. 100 mM ATP but in the absence of Mg 21 to prevent
- Published
- 2000
- Full Text
- View/download PDF
38. A multivalent peptide library approach identifies a novel Shiga toxin inhibitor that induces aberrant cellular transport of the toxin
- Author
-
Kiyotaka Nishikawa, Miho Watanabe, Eiji Kita, Katsura Igai, Kazumi Omata, Michael B. Yaffe, and Yasuhiro Natori
- Subjects
Protein Conformation ,Pentamer ,Golgi Apparatus ,Peptide ,Endoplasmic Reticulum ,medicine.disease_cause ,Biochemistry ,Shiga Toxin ,Mice ,symbols.namesake ,fluids and secretions ,Peptide Library ,hemic and lymphatic diseases ,Chlorocebus aethiops ,Genetics ,medicine ,Animals ,Amino Acid Sequence ,Peptide library ,Vero Cells ,Molecular Biology ,Escherichia coli ,Escherichia coli Infections ,chemistry.chemical_classification ,biology ,Toxin ,Endoplasmic reticulum ,Shiga toxin ,Golgi apparatus ,Molecular biology ,Mice, Inbred C57BL ,chemistry ,Mutation ,biology.protein ,symbols ,bacteria ,Female ,Peptides ,Biotechnology - Abstract
Infection with Shiga toxin (Stx)-producing Escherichia coli O157:H7 causes bloody diarrhea and hemorrhagic colitis in humans, sometimes resulting in fatal systemic complications. Among the known Stx family members, Stx2 is responsible for the most severe forms of disease. Stx2 binds to target cells via multivalent interactions between its B-subunit pentamer and globotriaosyl ceramide. After binding, it is first retrogradely transported to the Golgi and then to the endoplasmic reticulum (ER). Using a multivalent peptide library approach, we identified a tetravalent peptide that exhibits a high affinity for the Stx2 B-subunit pentamer (KD = 0.13 microM) and markedly inhibits Stx2 cytotoxicity. The tetravalent peptide exerted its inhibitory effects by inducing aberrant cellular transport of Stx2. Although the tetravalent peptide/Stx2 complex was incorporated into cells and translocated to the Golgi, this process was followed by the effective degradation of Stx2 in an acidic compartment rather than by its transfer to the ER. This peptide thoroughly protected mice from a fatal dose of E. coli O157:H7 even when administered after an established infection. Thus, the multivalent peptide library approach enabled the identification of a peptide-based Stx2 inhibitor that has remarkable therapeutic potency and appears to function by inducing aberrant cellular transport and degradation of Stx2.
- Published
- 2006
- Full Text
- View/download PDF
39. Activation of Phorbol Ester Responsive Form of Protein Kinase Cζ in Association with Ca2+-Induced Differentiation of Primary Cultured Mouse Epidermal Cells
- Author
-
Kiyotaka Nishikawa, Satoshi Yamamoto, Haruna Nagumo, and Ryuichi Kato
- Subjects
Pharmacology - Published
- 1997
- Full Text
- View/download PDF
40. Determination of the Specific Substrate Sequence Motifs of Protein Kinase C Isozymes
- Author
-
Lewis C. Cantley, Kiyotaka Nishikawa, Franz-Josef Johannes, Zhou Songyang, and Alex Toker
- Subjects
Protein Kinase C-alpha ,Stereochemistry ,Molecular Sequence Data ,Protein Kinase C beta ,Biology ,Biochemistry ,Isozyme ,Substrate Specificity ,Structure-Activity Relationship ,Humans ,Amino Acid Sequence ,Peptide library ,Molecular Biology ,Peptide sequence ,Protein Kinase C ,Protein kinase C ,chemistry.chemical_classification ,Cell Biology ,Amino acid ,Isoenzymes ,Intracellular signal transduction ,Kinetics ,Protein Kinase C-delta ,chemistry ,Sequence motif - Abstract
Protein kinase C (PKC) family members play significant roles in a variety of intracellular signal transduction processes, but information about the substrate specificities of each PKC family member is quite limited. In this study, we have determined the optimal peptide substrate sequence for each of nine human PKC isozymes (alpha, betaI, betaII, gamma, delta, epsilon, eta, mu, and zeta) by using an oriented peptide library. All PKC isozymes preferentially phosphorylated peptides with hydrophobic amino acids at position +1 carboxyl-terminal of the phosphorylated Ser and basic residues at position -3. All isozymes, except PKC mu, selected peptides with basic amino acids at positions -6, -4, and -2. PKC alpha, -betaI, -betaII, -gamma, and -eta selected peptides with basic amino acid at positions +2, +3, and +4, but PKC delta, -epsilon, -zeta, and -mu preferred peptides with hydrophobic amino acid at these positions. At position -5, the selectivity was quite different among the various isozymes; PKC alpha, -gamma, and -delta selected peptides with Arg at this position while other PKC isozymes selected hydrophobic amino acids such as Phe, Leu, or Val. Interestingly, PKC mu showed extreme selectivity for peptides with Leu at this position. The predicted optimal sequences from position -3 to +2 for PKC alpha, -betaI, -betaII, -gamma, -delta, and -eta were very similar to the endogenous pseudosubstrate sequences of these PKC isozymes, indicating that these core regions may be important to the binding of corresponding substrate peptides. Synthetic peptides based on the predicted optimal sequences for PKC alpha, -betaI,-delta, -zeta, and -mu were prepared and used for the determination of Km and Vmax for these isozymes. As judged by Vmax/Km values, these peptides were in general better substrates of the corresponding isozymes than those of the other PKC isozymes, supporting the idea that individual PKC isozymes have distinct optimal substrates. The structural basis for the selectivity of PKC isozymes is discussed based on residues predicted to form the catalytic cleft.
- Published
- 1997
- Full Text
- View/download PDF
41. JNK1/2-dependent phosphorylation of angulin-1/LSR is required for the exclusive localization of angulin-1/LSR and tricellulin at tricellular contacts in EpH4 epithelial sheet
- Author
-
Masayuki Murata, Daiki Nakatsu, Yuki Taguchi, Yukako Oda, Fumi Kano, Takashi Nishizono, Mikio Furuse, Kiyotaka Nishikawa, and Taichi Sugawara
- Subjects
Small interfering RNA ,Biology ,Cell Line ,Tight Junctions ,Serine ,Mice ,RNA interference ,Nitriles ,Genetics ,Animals ,Mitogen-Activated Protein Kinase 9 ,Mitogen-Activated Protein Kinase 8 ,Apigenin ,Phosphorylation ,Receptors, Lipoprotein ,Alanine ,Anthracenes ,Tight junction ,Kinase ,Epithelial Cells ,Cell Biology ,Tyrphostins ,Transmembrane protein ,Cell biology ,MARVEL Domain Containing 2 Protein ,Biochemistry - Abstract
Tricellular tight junctions (tTJs) are specialized structural variants of tight junctions within tricellular contacts of an epithelial sheet and comprise several transmembrane proteins including lipolysis-stimulated lipoprotein receptor (angulin-1/LSR) and tricellulin. To elucidate the mechanism of its formation, we carried out stepwise screening of kinase inhibitors followed by RNAi screening to identify kinases that regulate intracellular localization of angulin-1/LSR to the tTJs using a fluorescence image-based screen. We found that the activity of JNK1 and JNK2, but not JNK3, was required for the exclusive localization of angulin-1/LSR at the tTJs. Based on a bioinformatics approach, we estimated the potential phosphorylation site of angulin-1/LSR by JNK1 to be serine 288 and experimentally confirmed that JNK1 directly phosphorylates angulin-1/LSR at this site. We found that JNK2 was also involved in the phosphorylation of angulin-1/LSR. Furthermore, GFP-tagged angulin-1/LSR(S288A), in which serine 288 was substituted by alanine, was observed to be dispersed to bicellular junctions, indicating that phosphorylation of Ser288 is crucial for the exclusive localization of angulin-1/LSR and tricellulin at tTJs. Our fluorescence image-based screening for kinases inhibitor or siRNAs combined with the phosphorylation site prediction could become a versatile and useful tool to elucidate the mechanisms underlying the maintenance of tTJs regulated by kinase networks.
- Published
- 2013
42. Substrate ectodomain is critical for substrate preference and inhibition of γ-secretase
- Author
-
Seiko Ishihara, Mika Nobuhara, Toru Sasaki, Nobuto Kakuda, Kiyotaka Nishikawa, Takaomi C. Saido, Takashi Saito, Miho Watanabe-Takahashi, Satoru Funamoto, Yasuo Ihara, Tomohiro Miyasaka, and Masaki Nakano
- Subjects
Male ,Sialyltransferase ,medicine.medical_treatment ,Molecular Sequence Data ,General Physics and Astronomy ,Gene Expression ,Plasma protein binding ,CHO Cells ,General Biochemistry, Genetics and Molecular Biology ,Article ,Substrate Specificity ,Amyloid beta-Protein Precursor ,Mice ,Cricetulus ,Alzheimer Disease ,Antigens, CD ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Binding site ,Peptide sequence ,Multidisciplinary ,Protease ,Binding Sites ,biology ,Brain ,General Chemistry ,biology.organism_classification ,Recombinant Proteins ,Sialyltransferases ,HEK293 Cells ,Ectodomain ,Biochemistry ,biology.protein ,Amyloid Precursor Protein Secretases ,Peptides ,Amyloid precursor protein secretase ,Injections, Intraperitoneal ,Protein Binding - Abstract
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., γ-Secretase inhibitors are studied for their potential to treat Alzheimer’s disease, but their use is limited by side effects. Funamoto et al. show that γ-secretase preferentially cleaves substrates with short ectodomains and that inhibitors based on these ectodomains reduce disease-like pathology in mice.
- Published
- 2013
43. Identification of a Peptide-Based Neutralizer That Potently Inhibits Both Shiga Toxins 1 and 2 by Targeting Specific Receptor-Binding Regions
- Author
-
Kiyotaka Nishikawa, Eiji Kita, Miho Watanabe-Takahashi, Kazue Tsutsuki, and Yasuaki Takenaka
- Subjects
Cell Survival ,Immunology ,Peptide ,Enzyme-Linked Immunosorbent Assay ,medicine.disease_cause ,Endoplasmic Reticulum ,Escherichia coli O157 ,Shiga Toxin 1 ,Microbiology ,Shiga Toxin 2 ,Virulence factor ,Mice ,fluids and secretions ,Peptide Library ,Chlorocebus aethiops ,medicine ,Animals ,Peptide library ,Escherichia coli ,Vero Cells ,Escherichia coli Infections ,chemistry.chemical_classification ,biology ,Toxin ,Endoplasmic reticulum ,Shiga toxin ,Bacterial Infections ,biochemical phenomena, metabolism, and nutrition ,Molecular biology ,Specific Pathogen-Free Organisms ,Vesicular transport protein ,Mice, Inbred C57BL ,Protein Subunits ,Infectious Diseases ,chemistry ,Amino Acid Substitution ,biology.protein ,Parasitology ,Female ,Peptides - 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.
- Published
- 2013
44. The presence of phorbol ester responsive and non-responsive forms of the ζ isozyme of protein kinase C in mouse epidermal cells
- Author
-
Satoshi Yamamoto, Kouichi Maruyama, Ryuichi Kato, Kiyotaka Nishikawa, and Haruna Nagumo
- Subjects
Immunoprecipitation ,Blotting, Western ,Molecular Sequence Data ,Down-Regulation ,Biology ,Mice ,chemistry.chemical_compound ,Western blot ,medicine ,Animals ,Amino Acid Sequence ,Phosphorylation ,Kinase activity ,Protein kinase A ,Cells, Cultured ,Protein Kinase C ,Protein kinase C ,medicine.diagnostic_test ,Cell Biology ,Precipitin Tests ,Molecular biology ,Isoenzymes ,Molecular Weight ,Epidermal Cells ,Biochemistry ,chemistry ,Phorbol ,Tetradecanoylphorbol Acetate ,Electrophoresis, Polyacrylamide Gel ,Epidermis ,Signal transduction ,Signal Transduction - Abstract
The possible involvement of zeta isozyme of protein kinase C (PKC zeta) in phorbol ester-induced signal transduction was investigated in mouse epidermal cells. Western blot analysis of RESOURCE Q column chromatography eluates obtained from 105,000 g supernatants and particulate fractions of epidermal cells was performed using anti-PKC zeta specific antibody. Anti-PKC zeta antibody recognised proteins in low salt range corresponding to 25-125 mM NaCl (low salt-eluted PKC zeta; 1-PKC zeta) as well as high salt range corresponding to 175-300 mM NaCl (high salt-eluted PKC zeta; h-PKC zeta) in both subcellular fractions. 1-PKC zeta and h-PKC zeta were detected as a doublet protein of 79,000 and 85,000 M(r) in 105,000 g supernatants, but as a 79,000 M(r) protein in particulate fractions. Immunoprecipitated 1-PKC zeta and h-PKC zeta with anti-PKC zeta specific antibody possessed phosphatidylserine (PS)-dependent protein kinase activity, but neither 1-PKC zeta nor h-PKC zeta were further activated by 40 nM phorbol 12-myristate 13-acetate (PMA) in the presence of PS. Furthermore, 1-PKC zeta and h-PKC zeta can be autophosphorylated, indicating that both 1-PKC zeta and h-PKC zeta are PKC zeta. Treatment of intact epidermal cells with PMA or other PKC activators caused the apparent shift of 79,000 M(r) 1-PKC zeta to the 85,000 M(r) from in particulate fractions. Prolonged treatment of the cells with PMA induced the downregulation of both forms of 1-PKC zeta in particulate fractions. Under the same condition, 1-PKC zeta in 105,000 g supernatants and h-PKC zeta in both fractions did not respond to PMA. This apparent shift was reversible and the content ratio of 85,000 to 75,000 M(r) 1-PKC zeta was decreased by acid phosphatase treatment, indicating that the apparent shift results at least in part from phosphorylation of 79,000 M(r) 1-PKC zeta. Total activity of 1-PKC zeta was increased in association with the apparent shift from the 79,000 to 85,000 M(r) form in response to PMA treatment of intact epidermal cells. All of these results indicate that PKC zeta is present as multiple forms in mouse epidermal cells, and that especially 1-PKC zeta in particulate fractions play a significant role(s) in PMA-induced signal transduction in mouse epidermal cells.
- Published
- 1995
- Full Text
- View/download PDF
45. Intracellular phosphatidylserine is essential for retrograde membrane traffic through endosomes
- Author
-
David Sheff, Eiji Miyoshi, Shun-ichiro Iemura, Takatoshi Nakagawa, Wayne I. Lencer, Kiyotaka Nishikawa, Yasuo Uchiyama, Tohru Natsume, Kojiro Mukai, Soichi Wakatsuki, Naoyuki Taniguchi, Ryuichi Kato, Ryusuke Kuwahara, Yasunori Uchida, Daniel J.-F. Chinnapen, Hiroyuki Arai, Takao Inoue, Seiji Okazaki, Tomohiko Taguchi, Junya Hasegawa, Masato Koike, and Ryo Misaki
- Subjects
Endosome ,Endocytic cycle ,Green Fluorescent Proteins ,Golgi Apparatus ,Endosomes ,Phosphatidylserines ,Biology ,Endocytosis ,Crystallography, X-Ray ,Models, Biological ,symbols.namesake ,Chlorocebus aethiops ,Animals ,Humans ,Microscopy, Immunoelectron ,Vero Cells ,Multidisciplinary ,Membrane Proteins ,Biological membrane ,Intracellular Membranes ,Golgi apparatus ,Biological Sciences ,Transport protein ,Cell biology ,Protein Structure, Tertiary ,Pleckstrin homology domain ,Protein Transport ,Microscopy, Fluorescence ,COS Cells ,symbols ,RNA Interference ,Intracellular ,HeLa Cells ,Protein Binding - Abstract
Phosphatidylserine (PS) is a relatively minor constituent of biological membranes. Despite its low abundance, PS in the plasma membrane (PM) plays key roles in various phenomena such as the coagulation cascade, clearance of apoptotic cells, and recruitment of signaling molecules. PS also localizes in endocytic organelles, but how this relates to its cellular functions remains unknown. Here we report that PS is essential for retrograde membrane traffic at recycling endosomes (REs). PS was most concentrated in REs among intracellular organelles, and evectin-2 (evt-2), a protein of previously unknown function, was targeted to REs by the binding of its pleckstrin homology (PH) domain to PS. X-ray analysis supported the specificity of the binding of PS to the PH domain. Depletion of evt-2 or masking of intracellular PS suppressed membrane traffic from REs to the Golgi. These findings uncover the molecular basis that controls the RE-to-Golgi transport and identify a unique PH domain that specifically recognizes PS but not polyphosphoinositides.
- Published
- 2011
46. Recent progress of Shiga toxin neutralizer for treatment of infections by Shiga toxin-producing Escherichia coli
- Author
-
Kiyotaka Nishikawa
- Subjects
Polymers ,Virulence Factors ,Immunology ,Biology ,medicine.disease_cause ,Endoplasmic Reticulum ,Escherichia coli O157 ,Shiga Toxin 1 ,Shiga Toxin 2 ,Virulence factor ,Microbiology ,Mice ,medicine ,Immunology and Allergy ,Animals ,Combinatorial Chemistry Techniques ,Humans ,Receptor ,Peptide library ,Escherichia coli ,Escherichia coli Infections ,Binding Sites ,Globosides ,Toxin ,Trihexosylceramides ,Shiga toxin ,General Medicine ,Silanes ,Virology ,Anti-Bacterial Agents ,Diarrhea ,Serum Amyloid P-Component ,Infectious disease (medical specialty) ,Drug Design ,Hemolytic-Uremic Syndrome ,biology.protein ,Macrophages, Peritoneal ,Rabbits ,medicine.symptom ,Peptides ,Trisaccharides - Abstract
Infection with Shiga toxin (Stx)-producing Escherichia coli (STEC), including O157:H7, causes bloody diarrhea and hemorrhagic colitis in humans, occasionally resulting in fatal systemic complications, such as neurological damage and hemolytic-uremic syndrome. Because Stx is a major virulence factor of the infectious disease, a series of Shiga toxin neutralizers with various structural characteristics has been developed as promising therapeutic agents. Most of these agents function to bind to the toxin directly and inhibit the binding to its receptor present on the target cells. Other neutralizers do not inhibit receptor binding but induce aberrant intracellular transport of the toxin, resulting in effective detoxification. Such a novel type of Stx neutralizer provides a new therapeutic strategy against STEC infections. Here, recent progress of the development of Stx neutralizers is reviewed.
- Published
- 2010
47. Transmembrane BAX Inhibitor Motif Containing (TMBIM) Family Proteins Perturbs a trans-Golgi Network Enzyme, Gb3 Synthase, and Reduces Gb3 Biosynthesis*
- Author
-
Kiyotaka Nishikawa, Toshiyuki Yamaji, and Kentaro Hanada
- Subjects
Cell Survival ,Blotting, Western ,Green Fluorescent Proteins ,Molecular Sequence Data ,Globotriaosylceramide ,Drug Resistance ,Glycobiology and Extracellular Matrices ,Gene Expression ,Apoptosis ,Shiga Toxin 1 ,Transfection ,Biochemistry ,Receptors, N-Methyl-D-Aspartate ,Lactosylceramide ,chemistry.chemical_compound ,symbols.namesake ,Biosynthesis ,Cell Line, Tumor ,Humans ,Amino Acid Sequence ,music ,Molecular Biology ,music.instrument ,biology ,ATP synthase ,Base Sequence ,Globosides ,Reverse Transcriptase Polymerase Chain Reaction ,Trihexosylceramides ,Membrane Proteins ,Shiga toxin ,Cell Biology ,Golgi apparatus ,Galactosyltransferases ,Molecular biology ,Transmembrane protein ,chemistry ,Microscopy, Fluorescence ,Expression cloning ,biology.protein ,symbols ,Apoptosis Regulatory Proteins ,HeLa Cells ,Protein Binding ,trans-Golgi Network - Abstract
Globotriaosylceramide (Gb3) is a well known receptor for Shiga toxin (Stx), produced by enterohemorrhagic Escherichia coli and Shigella dysenteriae. The expression of Gb3 also affects several diseases, including cancer metastasis and Fabry disease, which prompted us to look for factors involved in its metabolism. In the present study, we isolated two cDNAs that conferred resistance to Stx-induced cell death in HeLa cells by expression cloning: ganglioside GM3 synthase and the COOH terminus region of glutamate receptor, ionotropic, N-methyl-d-asparate-associated protein 1 (GRINA), a member of the transmembrane BAX inhibitor motif containing (TMBIM) family. Overexpression of the truncated form, named GRINA-C, and some members of the full-length TMBIM family, including FAS inhibitory molecule 2 (FAIM2), reduced Gb3, and lactosylceramide was accumulated instead. The change of glycolipid composition was restored by overexpression of Gb3 synthase, suggesting that the synthase is affected by GRINA-C and FAIM2. Interestingly, the mRNA level of Gb3 synthase was unchanged. Rather, localization of the synthase as well as TGN46, a trans-Golgi network marker, was perturbed to form punctate structures, and degradation of the synthase in lysosomes was enhanced. Furthermore, GRINA-C was associated with Gb3 synthase. These observations may demonstrate a new type of posttranscriptional regulation of glycosyltransferases.
- Published
- 2010
48. Characterization of endogenous substrates for novel-type protein kinase C as well as conventional-type protein kinase C in primary cultured mouse epidermal cells
- Author
-
Kiyotaka Nishikawa, Chino Otsuka, Satoshi Yamamoto, and Ryuichi Kato
- Subjects
Biology ,Phosphoamino acid analysis ,Substrate Specificity ,Mice ,chemistry.chemical_compound ,Alkaloids ,medicine ,Animals ,Staurosporine ,Amino Acids ,Phosphorylation ,Protein kinase A ,Cells, Cultured ,Protein Kinase C ,Protein kinase C ,Skin ,Cell Biology ,Phosphoproteins ,Molecular biology ,Cell biology ,chemistry ,Cell culture ,Phorbol ,Tetradecanoylphorbol Acetate ,Signal transduction ,Signal Transduction ,Subcellular Fractions ,medicine.drug - Abstract
In primary cultured mouse epidermal cells, phorbol 12-myristate 13-acetate (PMA), which activates protein kinase C (PKC), induced changes in the phosphorylation levels of 10 proteins, termed KP-1 to 10, in two-dimensional PAGE. Seven of these proteins were phosphorylated and three were dephosphorylated. Similar changes were induced by other PKC activators, but not by inactive phorbol ester. Among these substrate proteins, phosphorylation of three proteins, i.e. KP-1 (pI 4.7/23,000 M r ), KP-2 (pI 4.7/20,700 M r ) and KP-10 (pI 4.7/25,000 M r was markedly enhanced by PMA and inhibited by a potent PKC inhibitor staurosporine. In vitro phosphorylation studies and phosphoamino acid analysis, using these proteins as substrate and PKC preparations obtained from epidermal cell lysate, revealed that KP-1 and -2 were directly phosphorylated by Ca 2+ -, phospholipid-dependent protein kinase (conventional-type PKC; cPKC), but not by Ca 2+ -independent, phospholipid-dependent protein kinase (novel-type PKC; nPKC). On the other hand, KP-10 was mainly phosphorylated by nPKC in intact epidermal cells. These results indicate that cPKC and nPKC in epidermal cells have different substrate specificity for endogenous proteins and may induce different signal transduction.
- Published
- 1992
- Full Text
- View/download PDF
49. Protein kinase C-dependent and -independent actions of a potent protein kinase C inhibitor, staurosporine
- Author
-
Hong Jiang, Ryuichi Kato, Kiyotaka Nishikawa, Motoko Ishihara, Satoshi Yamamoto, and Jian Chun Wang
- Subjects
genetic structures ,Indomethacin ,Down-Regulation ,Ornithine Decarboxylase ,12-O-Tetradecanoylphorbol-13-acetate ,Piperazines ,Ornithine decarboxylase ,Mice ,chemistry.chemical_compound ,Alkaloids ,1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine ,medicine ,Animals ,Staurosporine ,Phosphorylation ,Cells, Cultured ,Protein Kinase C ,Protein kinase C ,Pharmacology ,integumentary system ,biology ,Activator (genetics) ,fungi ,Proteins ,Isoquinolines ,Molecular biology ,Epidermal Cells ,chemistry ,Enzyme inhibitor ,Enzyme Induction ,Tetradecanoylphorbol Acetate ,biology.protein ,Electrophoresis, Polyacrylamide Gel ,Epidermis ,medicine.drug - Abstract
12-O-Tetradecanoylphorbol 13-acetate (TPA), an activator of protein kinase C (PKC), induced ornithine decarboxylase (ODC) in primary cultured mouse epidermal cells. Staurosporine, a potent protein kinase C inhibitor, also induced ODC activity. Both TPA- and staurosporine-caused ODC inductions were markedly suppressed in the PKC-down-regulated cells. Another PKC inhibitor, 1-(5-isoquinolinesulfonyl)-2-methylpiperazine (H-7), inhibited both TPA- and staurosporine-caused ODC inductions. H-7 by itself never induced ODC activity. Under our experimental conditions, staurosporine induced no detectable phosphorylation of endogenous proteins. TPA induced a translocation of PKC from cytosol to membrane whereas an optimal concentration of staurosporine to induce ODC did not induce an obvious translocation of PKC. Indomethacin, a cyclooxygenase inhibitor, inhibited staurosporine-caused ODC induction, but not TPA-caused ODC induction. Staurosporine induced specific morphological changes of epidermal cells both in normal and in PKC-down-regulated cells. These results indicate that staurosporine induces ODC activity in a PKC-dependent manner and morphological changes possibly through a PKC-independent mechanism. The mechanism of ODC induction caused by staurosporine may be in some way different from that caused by TPA.
- Published
- 1992
- Full Text
- View/download PDF
50. [Novel Shiga toxin inhibitor that induces aberrant cellular transport of the toxin]
- Author
-
Kiyotaka, Nishikawa
- Subjects
Peptide Library ,Drug Design ,Trihexosylceramides ,Amino Acid Motifs ,Animals ,Golgi Apparatus ,Humans ,Oligosaccharides ,Biological Transport ,Endoplasmic Reticulum ,Escherichia coli O157 ,Escherichia coli Infections ,Shiga Toxin - Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.