Network


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

Hotspot


Dive into the research topics where Koji Nishi is active.

Publication


Featured researches published by Koji Nishi.


Theranostics | 2012

Involvement of reactive oxygen species in sonodynamically induced apoptosis using a novel porphyrin derivative.

Nagahiko Yumita; Yumiko Iwase; Koji Nishi; Hajime Komatsu; Kazuyoshi Takeda; Kenji Onodera; Toshio Fukai; Toshihiko Ikeda; Shin-ichiro Umemura; Kazuho Okudaira; Yasunori Momose

In this study, we investigated the induction of apoptosis by ultrasound in the presence of the novel porphyrin derivative DCPH-P-Na(I). HL-60 cells were exposed to ultrasound for up to 3 min in the presence and absence of DCPH-P-Na(I), and the induction of apoptosis was examined by analyzing cell morphology, DNA fragmentation, and caspase-3 activity. Reactive oxygen species were measured by means of ESR and spin trapping technique. Cells treated with 8 μM DCPH-P-Na(I) and ultrasound clearly showed membrane blebbing and cell shrinkage, whereas significant morphologic changes were not observed in cells exposed to either ultrasound or DCPH-P-Na(I) alone. Also, DNA ladder formation and caspase-3 activation were observed in cells treated with both ultrasound and DCPH-P-Na(I) but not in cells treated with ultrasound or DCPH-P-Na(I) alone. In addition, the combination of DCPH-P-Na(I) and the same acoustical arrangement of ultrasound substantially enhanced nitroxide generation by the cells. Sonodynamically induced apoptosis, caspase-3 activation, and nitroxide generation were significantly suppressed by histidine. These results indicate that the combination of ultrasound and DCPH-P-Na(I) induced apoptosis in HL-60 cells. The significant reduction in sonodynamically induced apoptosis, nitroxide generation, and caspase-3 activation by histidine suggests active species such as singlet oxygen are important in the sonodynamic induction of apoptosis. These experimental results support the possibility of sonodynamic treatment for cancer using the induction of apoptosis.


Journal of Biological Chemistry | 2011

Structural Insights into Differences in Drug-binding Selectivity between Two Forms of Human α1-Acid Glycoprotein Genetic Variants, the A and F1*S Forms

Koji Nishi; Tomomi Ono; Teruya Nakamura; Naoko Fukunaga; Miyoko Izumi; Hiroshi Watanabe; Ayaka Suenaga; Toru Maruyama; Yuriko Yamagata; Stephen Curry; Masaki Otagiri

Human α1-acid glycoprotein (hAGP) in serum functions as a carrier of basic drugs. In most individuals, hAGP exists as a mixture of two genetic variants, the F1*S and A variants, which bind drugs with different selectivities. We prepared a mutant of the A variant, C149R, and showed that its drug-binding properties were indistinguishable from those of the wild type. In this study, we determined the crystal structures of this mutant hAGP alone and complexed with disopyramide (DSP), amitriptyline (AMT), and the nonspecific drug chlorpromazine (CPZ). The crystal structures revealed that the drug-binding pocket on the A variant is located within an eight-stranded β-barrel, similar to that found in the F1*S variant and other lipocalin family proteins. However, the binding region of the A variant is narrower than that of the F1*S variant. In the crystal structures of complexes with DSP and AMT, the two aromatic rings of each drug interact with Phe-49 and Phe-112 at the bottom of the binding pocket. Although the structure of CPZ is similar to those of DSP and AMT, its fused aromatic ring system, which is extended in length by the addition of a chlorine atom, appears to dictate an alternative mode of binding, which explains its nonselective binding to the F1*S and A variant hAGPs. Modeling experiments based on the co-crystal structures suggest that, in complexes of DSP, AMT, or CPZ with the F1*S variant, Phe-114 sterically hinders interactions with DSP and AMT, but not CPZ.


Biochimica et Biophysica Acta | 2002

Structural and drug-binding properties of α1-acid glycoprotein in reverse micelles

Koji Nishi; Norifumi Sakai; Yoshio Komine; Toru Maruyama; H. Brian Halsall; Masaki Otagiri

Abstract α 1 -Acid glycoprotein (AGP) is a glycoprotein that consists of 183 amino acid residues and five carbohydrate chains and binds to neutral and basic drugs. We examined the structural properties and ligand-binding capacity of AGP in interactions with reverse micelles. Also, detailed information was obtained by comparing several different states of AGP. Interaction with reverse micelles induced a unique conformational transition (β-sheet to α-helices) in AGP and decreased the binding capacity for the basic drug, chlorpromazine and the steroid hormone, progesterone to AGP. These structural conformations are very similar to those observed under conditions of acidity and high ionic strength (pH 2.0, 1.5 M NaCl). This structure seems to be an intermediate between the native state and the denatured state, possibly a molten globule. The present results suggest that when AGP interacts with the biomembrane, it undergoes a structural transition to a unique structure that differs from the native and denatured states and has a reduced ligand-binding capacity.


Drug Metabolism and Disposition | 2004

Construction of expression system for human α1-acid glycoprotein in Pichia pastoris and evaluation of its drug-binding properties

Koji Nishi; Naoko Fukunaga; Masaki Otagiri

Human α1-acid glycoprotein (hAGP) is a plasma glycoprotein that functions as a major carrier of basic ligands. This is the first report of the recombinant hAGP (rhAGP). In this study, rhAGP was expressed in the methylotropic yeast Pichia pastoris (GS115) using the expression vector, pPIC9, and then purified by anionic exchange, hydrophobic interaction, and gel filtration chromatography. The molecular weight of rhAGP was much lower than that of hAGP, because of the difference in glycan chain content. Results of glycopeptidase F digestion suggest that the peptide moiety of rhAGP was the same as that of hAGP. The results of circular dichroism spectra measurement indicated that rhAGP predominantly formed a β-sheet-rich structure that was the same as that of hAGP and typical of the lipocalin family. From the experiments using AGP-binding drugs (chlorpromazine, warfarin, and progesterone) and quinaldine red as a probe for the binding site, it was indicated that rhAGP also had the same ligand-binding capacity and binding site structure as hAGP. These findings strongly suggest that this recombinant hAGP (rhAGP) is very useful for the exploration of the ligand-binding site and biological function of hAGP.


Journal of Pharmaceutical Sciences | 2009

A site-directed mutagenesis study of drug-binding selectivity in genetic variants of human α1-acid glycoprotein

Koji Nishi; Megumi Ueno; Yuka Murakami; Naoko Fukunaga; Teruo Akuta; Daisuke Kadowaki; Hiroshi Watanabe; Ayaka Suenaga; Toru Maruyama; Masaki Otagiri

Human alpha(1)-acid glycoprotein (AGP), a major carrier of many basic drugs in circulation, consists of at least two genetic variants, namely A and F1*S variant. Interestingly, the variants of AGP have different drug-binding properties. The purpose of this study was to identify the amino acid residues that are responsible for the selectivity of drug binding to genetic variants of AGP using site-directed mutagenesis. First, we screened amino acid residues in the region proximal to position 100 that are involved in binding of warfarin and dipyridamole, which are F1*S-specific ligands, and of propafenone, which is an A-specific ligand, using ultrafiltration. In the F1*S variant, His97, His100, and Trp122 were involved in either warfarin- or dipyridamole-binding, while Glu92, His100, and Trp122 participated in the binding of propafenone in the A variant. Exchange of the residue at position 92 between AGP variants reversed the relative strength of propafenone binding to the two variants, but had a markedly different effect on binding of warfarin and dipyridamole. These findings indicate that the amino acid residue at position 92 plays a significant role in drug-binding selectivity in AGP variants, especially for drugs that preferentially bind to the A variant.


Pharmaceutical Research | 2004

Tryptophan Residues Play an Important Role in the Extraordinarily High Affinity Binding Interaction of UCN-01 to Human α-1-Acid Glycoprotein

Masaaki Katsuki; Victor Tuan Giam Chuang; Koji Nishi; Ayaka Suenaga; Masaki Otagiri

AbstractPurpose. To investigate the factors that contribute to the exceptionally high affinity binding of UCN-01 to human α1-acid glycoprotein (hAGP). Methods. Interactions between UCN-01, UCN-02, and staurosporine with native and chemically modified hAGPs were examined using ultracentrifugation and spectroscopic analysis. Results. The binding affinity of staurosporine, as well as UCN-02, to hAGP was lower than that of UCN-01 by 20- and 100-fold respectively. The percentage of UCN-01 that binds to hAGP was low at acidic pH but increased with increasing pH, reaching a maximum at pH 7.4. The binding of UCN-01 to desialylated hAGP was comparable to that of hAGP. No significant difference was found for the binding of UCN-01 to F1*S and A variants of hAGP. Chemical modification of the His, Lys, Trp, and Tyr residues caused a decrease in percentage of bound UCN-01. Trp-modified hAGP showed the largest decrease in binding. Tryptophanyl fluorescence quenching results indicate that Trp residues play a prominent role in the binding of UCN-01 to hAGP. Conclusions. A substituent at position C-7 of UCN-01 appeared to influence the binding specificity of the drug, and Trp residues in hAGP play a prominent role in the high affinity binding of UCN-01 to hAGP.


Proteins | 2006

Involvement of disulfide bonds and histidine 172 in a unique β‐sheet to α‐helix transition of α1‐acid glycoprotein at the biomembrane interface

Koji Nishi; Yoshio Komine; Naoko Fukunaga; Toru Maruyama; Ayaka Suenaga; Masaki Otagiri

Human α1‐acid glycoprotein (AGP), which is comprised of 183 amino acid residues and 5 carbohydrate chains, is a major plasma protein that binds to basic and neutral drugs as well as to steroid hormones. It has a β‐sheet–rich structure in aqueous solution. Our previous findings suggest that AGP forms an α‐helix structure through an interaction with biomembranes. We report herein on a study of the mechanism of α‐helix formation in AGP using various modified AGPs. The disulfide reduced AGP (R‐AGP) was extensively unfolded, whereas asialylated AGP (A‐AGP) maintained the native structure. Intriguingly, reduced and asialylated AGP (RA‐AGP) increased the α‐helix content as observed in the presence of biomembrane models, and showed a significant decrease in ligand binding capacity. This suggests that AGP has an innate tendency to form an α‐helix structure, and disulfide bonds are a key factor in the conformational transition between the β‐sheet and α‐helix structures. However, RA‐AGP with all histidine residues chemically modified (HRA‐AGP) was found to lose the intrinsic ability to form an α‐helix structure. Furthermore, disulfide reduction of the H172A mutant expressed in Pichia pastoris also caused a similar loss of folding ability. The present results indicate that disulfide bonds and the C‐terminal region, including H172 of AGP, play important roles in α‐helix formation in the interaction of the protein with biomembranes. Proteins 2006.


PLOS ONE | 2017

Long chain fatty acids alter the interactive binding of ligands to the two principal drug binding sites of human serum albumin

Keishi Yamasaki; Saya Hyodo; Kazuaki Taguchi; Koji Nishi; Noriyuki Yamaotsu; Shuichi Hirono; Victor Tuan Giam Chuang; Hakaru Seo; Toru Maruyama; Masaki Otagiri

A wide variety of drugs bind to human serum albumin (HSA) at its two principal sites, namely site I and site II. A number of reports indicate that drug binding to these two binding sites are not completely independent, and that interactions between ligands of these two discrete sites can play a role. In this study, the effect of the binding of long-chain fatty acids on the interactive binding between dansyl-L-asparagine (DNSA; site I ligand) and ibuprofen (site II ligand) at pH6.5 was examined. Binding experiments showed that the binding of sodium oleate (Ole) to HSA induces conformational changes in the molecule, which, in turn, changes the individual binding of DNSA and ibuprofen, as well as the mode of interaction between these two ligands from a ‘competitive-like’ allosteric interaction in the case of the defatted HSA conformer to a ‘nearly independent’ binding in the case of non-defatted HSA conformer. Circular dichroism measurements indicated that ibuprofen and Ole are likely to modify the spatial orientation of DNSA at its binding site. Docking simulations suggest that the long-distance electric repulsion between DNSA and ibuprofen on defatted HSA contributes to a ‘competitive-like’ allosteric interaction, whereas extending the distance between ligands and/or increasing the flexibility or size of the DNSA binding site in fatted HSA evokes a change in the interaction mode to ‘nearly independent’ binding. The present findings provide further insights into the structural dynamics of HSA upon the binding of fatty acids, and its effects on drug binding and drug-drug interactions that occur on HSA.


FEBS Letters | 2005

Cooperative effect of hydrophobic and electrostatic forces on alcohol-induced α-helix formation of α1-acid glycoprotein

Koji Nishi; Yoshio Komine; Norifumi Sakai; Toru Maruyama; Masaki Otagiri

α1‐Acid glycoprotein (AGP) is a serum glycoprotein that mainly binds basic drugs. Previous reports have shown that AGP converts from a β‐sheet to an α‐helix upon interaction with biomembranes. In the current studies, we found that alkanols, diols, and halogenols all induce this conformational change. Increased length and bulkiness of the hydrocarbon group and the presence of a halogen atom promoted this conversion, whereas the presence of a hydroxyl group inhibited it. Moreover, the effect was dependent on the hydrophobic and electrostatic properties of the alcohols. These results indicate that, in a membrane environment, hydrophobic and electrostatic factors cooperatively induce the transition of AGP from a β‐sheet to an α‐helix.


Journal of Pharmaceutical Sciences | 2012

Characterization of Hepatic Cellular Uptake of α1-Acid Glycoprotein (AGP), Part 2: Involvement of Hemoglobin β-Chain on Plasma Membranes in the Uptake of Human AGP by Liver Parenchymal Cells

Hisakazu Komori; Koji Nishi; Nao Uehara; Hiroshi Watanabe; Tsuyoshi Shuto; Ayaka Suenaga; Toru Maruyama; Masaki Otagiri

Human α(1) -acid glycoprotein (AGP), a lipocalin family member, serves as a carrier for basic drugs and endogenous hormones. It is mainly distributed in the liver and also has anti-inflammatory effects. We previously discovered a protein in liver parenchymal cells that interacts with AGP and it was identified as hemoglobin β-chain (HBB). The purpose of this study was to clarify the role of HBB in the hepatic cellular uptake of AGP. Ligand blotting experiments showed that the interaction of (125) I-AGP with hemoglobin was saturable and was significantly suppressed in the presence of excess unlabeled AGP. In addition, the cellular uptake of fluorescein isothiocianate-AGP by HepG2 cells was saturable and temperature dependent. This uptake was inhibited by fillipin and methyl-β-cyclodextrin, but not chlorpromazine, suggesting that AGP is taken up via caveolae/lipid rafts endocytic pathway. Immunostaining showed that HBB and caveolin-1, exclusively expressed in caveolae, were partially colocalized on the plasma membranes of HepG2 cells. HBB knockdown with siRNA decreased the uptake of AGP by HepG2 cells by 40%, and exogenous hemoglobin inhibited the uptake by 40%-50%. These findings indicate that HBB is located on the liver plasma membrane and that it contributes to the intracellular uptake of AGP.

Collaboration


Dive into the Koji Nishi's collaboration.

Top Co-Authors

Avatar

Masaki Otagiri

National University of Malaysia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nagahiko Yumita

Yokohama College of Pharmacy

View shared research outputs
Top Co-Authors

Avatar

Yumiko Iwase

Yokohama College of Pharmacy

View shared research outputs
Top Co-Authors

Avatar

Toshihiko Ikeda

Yokohama College of Pharmacy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge