Network


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

Hotspot


Dive into the research topics where Takashi Okura is active.

Publication


Featured researches published by Takashi Okura.


Drug Metabolism and Disposition | 2008

Involvement of the Pyrilamine Transporter, a Putative Organic Cation Transporter, in Blood-Brain Barrier Transport of Oxycodone

Takashi Okura; Asami Hattori; Yusuke Takano; Takenori Sato; Margareta Hammarlund-Udenaes; Tetsuya Terasaki; Yoshiharu Deguchi

The purpose of this study was to characterize blood-brain barrier (BBB) transport of oxycodone, a cationic opioid agonist, via the pyrilamine transporter, a putative organic cation transporter, using conditionally immortalized rat brain capillary endothelial cells (TR-BBB13). Oxycodone and [3H]pyrilamine were both transported into TR-BBB13 cells in a temperature- and concentration-dependent manner with Km values of 89 and 28 μM, respectively. The initial uptake of oxycodone was significantly enhanced by preloading with pyrilamine and vice versa. Furthermore, mutual uptake inhibition by oxycodone and pyrilamine suggests that a common mechanism is involved in their transport. Transport of both substrates was inhibited by type II cations (quinidine, verapamil, and amantadine), but not by classic organic cation transporter (OCT) substrates and/or inhibitors (tetraethylammonium, 1-methyl-4-phenylpyridinium, and corticosterone), substrates of OCTN1 (ergothioneine) and OCTN2 (l-carnitine), or organic anions. The transport was inhibited by metabolic inhibitors (rotenone and sodium azide) but was insensitive to extracellular sodium and membrane potential for both substrates. Furthermore, the transport of both substrates was increased at alkaline extracellular pH and decreased in the presence of a protonophore (carbonyl cyanide-p-trifluoromethoxyphenylhydrazone). Intracellular acidification induced with ammonium chloride enhanced the uptakes, suggesting that the transport is driven by an oppositely directed proton gradient. The brain uptake of oxycodone measured by in situ rat brain perfusion was increased in alkaline perfusate and was significantly inhibited by pyrilamine. These results suggest that blood-brain barrier transport of oxycodone is at least partly mediated by a common transporter with pyrilamine, and this transporter is an energy-dependent, proton-coupled antiporter.


Journal of Neurochemistry | 2002

Internalization of basic fibroblast growth factor at the mouse blood–brain barrier involves perlecan, a heparan sulfate proteoglycan

Yoshiharu Deguchi; Hiroshi Okutsu; Takashi Okura; Shizuo Yamada; Ryohei Kimura; Takuro Yuge; Akihiko Furukawa; Kazuhiro Morimoto; Masanori Tachikawa; Sumio Ohtsuki; Ken-ichi Hosoya; Tetsuya Terasaki

In this study, the internalization mechanism of basic fibroblast growth factor (bFGF) at the blood–brain barrier (BBB) was investigated using a conditionally immortalized mouse brain capillary endothelial cell line (TM‐BBB4 cells) as an in vitro model of the BBB and the corresponding receptor was identified using immunohistochemical analysis. The heparin‐resistant binding of [125I]bFGF to TM‐BBB4 cells was found to be time‐, temperature‐, osmolarity‐ and concentration‐dependent. Kinetic analysis of the cell‐surface binding of [125I]bFGF to TM‐BBB4 cells revealed saturable binding with a half‐saturation constant of 76 ± 24 nm and a maximal binding capacity of 183 ± 17 pmol/mg protein. The heparin‐resistant binding of [125I]bFGF to TM‐BBB4 was significantly inhibited by a cationic polypeptide poly‐L‐lysine (300 µm), and compounds which contain a sulfate moiety, e.g. heparin and chondroitin sulfate‐B (each 10 µg/mL). Moreover, the heparin‐resistant binding of [125I]bFGF in TM‐BBB4 cells was significantly reduced by 50% following treatment with sodium chlorate, suggesting the loss of perlecan (a core protein of heparan sulfate proteoglycan, HSPG) from the extracellular matrix of the cells. This type of binding is consistent with the involvement HSPG‐mediated endocytosis. RT‐PCR analysis revealed that HSPG mRNA and FGFR1 and FGFR2 (tyrosine‐kinase receptors for bFGF) mRNA are expressed in TM‐BBB4 cells. Moreover, immunohistochemical analysis demonstrated that perlecan is expressed on the abluminal membrane of the mouse brain capillary. These results suggest that bFGF is internalized via HSPG, which is expressed on the abluminal membrane of the BBB. HSPG at the BBB may play a role in maintaining the BBB function due to acceptance of the bFGF secreted from astrocytes.


Journal of Pharmaceutical Sciences | 2011

Diphenhydramine Active Uptake at the Blood–Brain Barrier and Its Interaction with Oxycodone in vitro and in Vivo

Muhammad Waqas Sadiq; Annika Borgs; Takashi Okura; Keita Shimomura; Sayaka Kato; Yoshiharu Deguchi; Britt Jansson; Sven Björkman; Tetsuya Terasaki; Margareta Hammarlund-Udenaes

Diphenhydramine (DPHM) and oxycodone are weak bases that are able to form cations. Both drugs show active uptake at the blood-brain barrier (BBB). There is thus a possibility for a pharmacokinetic interaction between them by competition for the same uptake transport system. The experiments of the present study were designed to study the transport of DPHM across the BBB and its interaction with oxycodone in vitro and in vivo. In vitro, the interaction between the drugs was studied using conditionally immortalized rat brain capillary endothelial cells (TR-BBB13 cells). The in vivo relevance of the in vitro findings was studied in rats using brain and blood microdialysis. DPHM was actively transported across the BBB in vitro (TR-BBB13 cells). Oxycodone competitively inhibited DPHM uptake with a K(i) value of 106 μM. DPHM also competitively inhibited oxycodone uptake with a K(i) value of 34.7 μM. In rats, DPHM showed fivefold higher unbound concentration in brain interstitial fluid (ISF) than in blood, confirming a net active uptake. There was no significant interaction between DPHM and oxycodone in vivo. This accords with the results of the in vitro experiments because the unbound plasma concentrations that could be attained in vivo, without causing adverse effects, were far below the K(i) values.


Molecular Cancer Therapeutics | 2007

Cell cycle phenotype-based optimization of G2-abrogating peptides yields CBP501 with a unique mechanism of action at the G2 checkpoint

Shi-Ken Sha; Takuji Sato; Hidetaka Kobayashi; Machiyo Ishigaki; Sayaka Yamamoto; Hitoshi Sato; Asako Takada; Sigetosi Nakajyo; Yasuo Mochizuki; Jonathan M. Friedman; Fong-Chi Cheng; Takashi Okura; Ryohei Kimura; Donald Kufe; Daniel D. VonHoff; Takumi Kawabe

Cell cycle G2 checkpoint abrogation is an attractive strategy for sensitizing cancer cells to DNA-damaging anticancer agent without increasing adverse effects on normal cells. However, there is no single proven molecular target for this therapeutic approach. High-throughput screening for molecules inhibiting CHK1, a kinase that is essential for the G2 checkpoint, has not yet yielded therapeutic G2 checkpoint inhibitors, and the tumor suppressor phenotypes of ATM and CHK2 suggest they may not be ideal targets. Here, we optimized two G2 checkpoint-abrogating peptides, TAT-S216 and TAT-S216A, based on their ability to reduce G2 phase accumulation of DNA-damaged cells without affecting M phase accumulation of cells treated with a microtubule-disrupting compound. This approach yielded a peptide CBP501, which has a unique, focused activity against molecules that phosphorylate Ser216 of CDC25C, including MAPKAP-K2, C-Tak1, and CHK1. CBP501 is >100-fold more potent than TAT-S216A and retains its selectivity for cancer cells. CBP501 is unusually stable, enters cells rapidly, and increases the cytotoxicity of DNA-damaging anticancer drugs against cancer cells without increasing adverse effects. These findings highlight the potency of CBP501 as a G2-abrogating drug candidate. This report also shows the usefulness of the cell cycle phenotype-based protocol for identifying G2 checkpoint-abrogating compounds as well as the potential of peptide-based compounds as focused multitarget inhibitors. [Mol Cancer Ther 2007;6(1):147–53]


Journal of Pharmaceutical Sciences | 2014

Transport characteristics of tramadol in the blood-brain barrier.

Atsushi Kitamura; Kei Higuchi; Takashi Okura; Yoshiharu Deguchi

Tramadol is a centrally acting analgesic whose action is mediated by both agonistic activity at opioid receptors and inhibitory activity on neuronal reuptake of monoamines. The purpose of this study was to characterize the blood-brain barrier (BBB) transport of tramadol by means of microdialysis studies in rat brain and in vitro studies with human immortalized brain capillary endothelial cells (hCMEC/D3). The Kp,uu,brain value of tramadol determined by rat brain microdialysis was greater than unity, indicating that tramadol is actively taken up into the brain across the BBB. Tramadol was transported into hCMEC/D3 cells in a concentration-dependent manner. The uptake was inhibited by type II cations (pyrilamine, verapamil, etc.), but not by substrates of organic cation transporter OCTs or OCTN2. It was also inhibited by a metabolic inhibitor but was independent of extracellular sodium or membrane potential. The uptake was altered by changes of extracellular pH, and by ammonium chloride-induced intracellular acidification, suggesting that transport of tramadol is driven by an oppositely directed proton gradient. Thus, our in vitro and in vivo results suggest that tramadol is actively transported, at least in part, from blood to the brain across the BBB by proton-coupled organic cation antiporter.


Journal of Pharmaceutical Sciences | 2011

Functional characterization of rat plasma membrane monoamine transporter in the blood–brain and blood–cerebrospinal fluid barriers

Takashi Okura; Sayaka Kato; Yusuke Takano; Takenori Sato; Atsushi Yamashita; Riyo Morimoto; Sumio Ohtsuki; Tetsuya Terasaki; Yoshiharu Deguchi

This study investigated the expression and functional roles of rat plasma membrane monoamine transporter (rPMAT) in the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier by using in vitro brain barrier model cells (TR-BBB13 and TR-CSFB3 cells) and multiple in vivo experimental techniques. Quantitative reverse transcription-polymerase chain reaction analysis showed relatively high expression of rPMAT mRNA in TR-BBB13 and TR-CSFB3 cells. 1-Methyl-4-phenylpyridinium (MPP(+) ) was transported into rPMAT-expressing cells in a sodium-independent manner. [(3) H]MPP(+) was taken up concentration dependently by TR-BBB13 and TR-CSFB3 cells with K(m) values similar to that of rPMAT-expressing cells. [(3) H]MPP(+) transports into these cells were markedly inhibited by serotonin, dopamine, and cationic drugs. rPMAT small interfering RNA (siRNA) significantly suppressed the [(3) H]MPP(+) uptake by TR-BBB13 cells. Intracerebrally injected [(3) H]MPP(+) was eliminated from the brain parenchymal region, whereas brain [(3) H]MPP(+) uptake did not increase with time during in situ brain perfusion, suggesting that the brain-to-blood transport across the BBB predominates over the blood-to-brain transport. Brain microdialysis studies revealed that the elimination across the BBB was significantly decreased by coperfusion of unlabelled MPP(+) , serotonin, or dopamine. [(3) H]MPP(+) was also eliminated from the CSF. These findings suggest that PMAT in brain barriers functions as the brain-to-blood transporter to regulate brain concentrations of organic cations including monoamines and cationic neurotoxins.


Fluids and Barriers of the CNS | 2013

Functional expression of a proton-coupled organic cation (H+/OC) antiporter in human brain capillary endothelial cell line hCMEC/D3, a human blood–brain barrier model

Keita Shimomura; Takashi Okura; Sayaka Kato; Pierre-Olivier Couraud; Jean-Michel Schermann; Tetsuya Terasaki; Yoshiharu Deguchi

BackgroundKnowledge of the molecular basis and transport function of the human blood–brain barrier (BBB) is important for not only understanding human cerebral physiology, but also development of new central nervous system (CNS)-acting drugs. However, few studies have been done using human brain capillary endothelial cells, because human brain materials are difficult to obtain. The purpose of this study is to clarify the functional expression of a proton-coupled organic cation (H+/OC) antiporter in human brain capillary endothelial cell line hCMEC/D3, which has been recently developed as an in vitro human BBB model.MethodsDiphenhydramine, [3H]pyrilamine and oxycodone were used as cationic drugs that proved to be H+/OC antiporter substrates. The in vitro uptake experiments by hCMEC/D3 cells were carried out under several conditions.ResultsDiphenhydramine and [3H]pyrilamine were both transported into hCMEC/D3 cells in a time- and concentration-dependent manner with Km values of 59 μM and 19 μM, respectively. Each inhibited uptake of the other in a competitive manner, suggesting that a common mechanism is involved in their transport. The diphenhydramine uptake was significantly inhibited by amantadine and quinidine, but not tetraethylammonium and 1-methyl-4-phenylpyridinium (substrates for well-known organic cation transporters). The uptake was inhibited by metabolic inhibitors, but was insensitive to extracellular sodium and membrane potential. Further, the uptake was increased by extracellular alkalization and intracellular acidification. These transport properties are completely consistent with those of previously characterized H+/OC antiporter in rat BBB.ConclusionsThe present results suggest that H+/OC antiporter is functionally expressed in hCMEC/D3 cells.


British Journal of Pharmacology | 2003

Different distribution of morphine and morphine-6β-glucuronide after intracerebroventricular injection in rats

Takashi Okura; Masanori Saito; Misato Nakanishi; Noriyuki Komiyama; Aki Fujii; Shizuo Yamada; Ryohei Kimura

We investigated the distribution of morphine and morphine‐6β‐glucuronide (M6G) in the brain and spinal cord after intracerebroventricular (i.c.v.) injection of each drug in rats. The cerebrospinal fluid (CSF) concentration of M6G was 5–37 times greater than that of morphine 10, 60 and 120 min after the i.c.v. injection. The apparent elimination clearance of M6G from the CSF was 10 times lower than that of morphine. The intrathecal CSF concentration of M6G measured by the microdialysis method was 29–79 times greater than that of morphine, and M6G was rapidly distributed into the intrathecal space after the i.c.v. injection. M6G was detected in the cerebrum, brainstem, cerebellum and spinal cord at concentrations 2–21 times higher than morphine after the i.c.v. injection of each drug. The distribution volume of M6G in rat brain slices was three times lower than that of morphine, and close to the extracellular fluid space in the brain regions corresponding to the vicinity of the opioid receptors. These brain distribution characteristics of M6G, namely, low clearance from the central nervous system, localization in the extracellular fluid and rapid distribution into the intrathecal space, may contribute to the potent analgesic effect of M6G after i.c.v. injection.


Journal of Pharmaceutical Sciences | 2010

Drug–drug interaction between oxycodone and adjuvant analgesics in blood–brain barrier transport and antinociceptive effect

Yusuke Nakazawa; Takashi Okura; Keita Shimomura; Tetsuya Terasaki; Yoshiharu Deguchi

To examine possible blood-brain barrier (BBB) transport interactions between oxycodone and adjuvant analgesics, we firstly screened various candidates in vitro using [(3)H]pyrilamine, a substrate of the oxycodone transporter, as a probe drug. The uptake of [(3)H]pyrilamine by conditionally immortalized rat brain capillary endothelial cells (TR-BBB13) was inhibited by antidepressants (amitriptyline, imipramine, clomipramine, amoxapine, and fluvoxamine), antiarrhythmics (mexiletine, lidocaine, and flecainide), and ketamine. On the other hand, antiepileptics (carbamazepine, phenytoin, and clonazepam) and corticosteroids (dexamethasone and prednisolone) did not inhibit [(3)H]pyrilamine uptake, with the exception of sodium valproate. The uptake of oxycodone was significantly inhibited in a concentration-dependent manner by amitriptyline, fluvoxamine and mexiletine with K(i) values of 13, 65, and 44 microM, respectively. These K(i) values are 5-300 times greater than the human therapeutic plasma concentrations. Finally, we evaluated in vivo interaction between oxycodone and amitriptyline in mice. Antinociceptive effects of oxycodone were increased by coadministration of amitriptyline. The oxycodone concentrations in plasma and brain were not changed by coadministration of amitriptyline. Overall, the results suggest that several adjuvant analgesics may interact with the BBB transport of oxycodone at relatively high concentrations. However, it is unlikely that there would be any significant interaction at therapeutically or pharmacologically relevant concentrations.


Journal of Pharmacy and Pharmacology | 2009

Effects of quinidine on antinociception and pharmacokinetics of morphine in rats.

Takashi Okura; Yuki Morita; Yoshihiko Ito; Yoshiyuki Kagawa; Shizuo Yamada

AimThe aim of this study was to investigate the effect of quinidine, a P‐glycoprotein inhibitor, on the pharmacokinetics and pharmacodynamics of morphine in rats.

Collaboration


Dive into the Takashi Okura's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge