Skip to main content
Top
Published in: Fluids and Barriers of the CNS 1/2018

Open Access 01-12-2018 | Research

Role of cationic drug-sensitive transport systems at the blood-cerebrospinal fluid barrier in para-tyramine elimination from rat brain

Authors: Shin-ichi Akanuma, Yuhei Yamazaki, Yoshiyuki Kubo, Ken-ichi Hosoya

Published in: Fluids and Barriers of the CNS | Issue 1/2018

Login to get access

Abstract

Background

para-Tyramine (p-TA) is a biogenic amine which is involved in multiple neuronal signal transductions. Since the concentration of p-TA in dog cerebrospinal fluid (CSF) has been reported to be greater than that in plasma, it is proposed that clearance of cerebral p-TA is important for normal function. The purpose of this study was to examine the role of the blood–brain barrier and blood-cerebrospinal fluid barrier (BCSFB) in p-TA clearance from the brain.

Methods

In vivo [3H]p-TA elimination from rat cerebral cortex and from CSF was examined after intracerebral and intracerebroventricular administration, respectively. To evaluate BCSFB-mediated p-TA transport, [3H]p-TA uptake by isolated rat choroid plexus and conditionally immortalized rat choroid plexus epithelial cells, TR-CSFB3 cells, was performed.

Results

The half-life of [3H]p-TA elimination from rat CSF was found to be 2.9 min, which is 62-fold faster than that from rat cerebral cortex. In addition, this [3H]p-TA elimination from the CSF was significantly inhibited by co-injection of excess unlabeled p-TA. Thus, carrier-mediated p-TA transport process(es) are assumed to take part in p-TA elimination from the CSF. Since it is known that transporters at the BCSFB participate in compound elimination from the CSF, [3H]p-TA transport in ex vivo and in vitro models of rat BCSFB was examined. The [3H]p-TA uptake by isolated rat choroid plexus and TR-CSFB3 cells was time-dependent and was inhibited by unlabeled p-TA, indicating carrier-mediated p-TA transport at the BCSFB. The p-TA uptake by isolated choroid plexus and TR-CSFB3 cells was not reduced in the absence of extracellular Na+ and Cl, and in the presence of substrates of typical organic cation transporters. However, this p-TA uptake was significantly inhibited by cationic drugs such as propranolol, imipramine, amantadine, verapamil, and pyrilamine. Moreover, p-TA uptake by TR-CSFB3 cells took place in an oppositely-directed H+ gradient manner. Therefore, this suggested that p-TA transport at the BCSFB involves cationic drug-sensitive transport systems which are distinct from typical plasma membrane organic cation transporters.

Conclusion

Our study indicates that p-TA elimination from the CSF is greater than that from the cerebral cortex. Moreover, it is suggested that cationic drug-sensitive transport systems in the BCSFB participate in this p-TA elimination from the CSF.
Literature
1.
go back to reference Borkum JM. Migraine triggers and oxidative stress: a narrative review and synthesis. Headache. 2016;56:12–35.CrossRefPubMed Borkum JM. Migraine triggers and oxidative stress: a narrative review and synthesis. Headache. 2016;56:12–35.CrossRefPubMed
2.
go back to reference Ledonne A, Berretta N, Davoli A, Rizzo GR, Bernardi G, Mercuri NB. Electrophysiological effects of trace amines on mesencephalic dopaminergic neurons. Front Syst Neurosci. 2011;5:56.CrossRefPubMedPubMedCentral Ledonne A, Berretta N, Davoli A, Rizzo GR, Bernardi G, Mercuri NB. Electrophysiological effects of trace amines on mesencephalic dopaminergic neurons. Front Syst Neurosci. 2011;5:56.CrossRefPubMedPubMedCentral
3.
go back to reference Youdim MB, Weinstock M. Therapeutic applications of selective and non-selective inhibitors of monoamine oxidase A and B that do not cause significant tyramine potentiation. Neurotoxicology. 2004;25:243–50.CrossRefPubMed Youdim MB, Weinstock M. Therapeutic applications of selective and non-selective inhibitors of monoamine oxidase A and B that do not cause significant tyramine potentiation. Neurotoxicology. 2004;25:243–50.CrossRefPubMed
4.
go back to reference Revel FG, Moreau JL, Gainetdinov RR, Bradaia A, Sotnikova TD, Mory R, et al. TAAR1 activation modulates monoaminergic neurotransmission, preventing hyperdopaminergic and hypoglutamatergic activity. Proc Natl Acad Sci USA. 2011;108:8485–90.CrossRefPubMedPubMedCentral Revel FG, Moreau JL, Gainetdinov RR, Bradaia A, Sotnikova TD, Mory R, et al. TAAR1 activation modulates monoaminergic neurotransmission, preventing hyperdopaminergic and hypoglutamatergic activity. Proc Natl Acad Sci USA. 2011;108:8485–90.CrossRefPubMedPubMedCentral
5.
go back to reference Revel FG, Moreau JL, Pouzet B, Mory R, Bradaia A, Buchy D, et al. A new perspective for schizophrenia: TAAR1 agonists reveal antipsychotic- and antidepressant-like activity, improve cognition and control body weight. Mol Psychiatry. 2013;18:543–56.CrossRefPubMed Revel FG, Moreau JL, Pouzet B, Mory R, Bradaia A, Buchy D, et al. A new perspective for schizophrenia: TAAR1 agonists reveal antipsychotic- and antidepressant-like activity, improve cognition and control body weight. Mol Psychiatry. 2013;18:543–56.CrossRefPubMed
6.
go back to reference Wolinsky TD, Swanson CJ, Smith KE, Zhong H, Borowsky B, Seeman P, et al. The Trace Amine 1 receptor knockout mouse: an animal model with relevance to schizophrenia. Genes Brain Behav. 2007;6:628–39.CrossRefPubMed Wolinsky TD, Swanson CJ, Smith KE, Zhong H, Borowsky B, Seeman P, et al. The Trace Amine 1 receptor knockout mouse: an animal model with relevance to schizophrenia. Genes Brain Behav. 2007;6:628–39.CrossRefPubMed
7.
go back to reference Lindemann L, Meyer CA, Jeanneau K, Bradaia A, Ozmen L, Bluethmann H, et al. Trace amine-associated receptor 1 modulates dopaminergic activity. J Pharmacol Exp Ther. 2008;324:948–56.CrossRefPubMed Lindemann L, Meyer CA, Jeanneau K, Bradaia A, Ozmen L, Bluethmann H, et al. Trace amine-associated receptor 1 modulates dopaminergic activity. J Pharmacol Exp Ther. 2008;324:948–56.CrossRefPubMed
8.
go back to reference Xie Z, Miller GM. Beta-phenylethylamine alters monoamine transporter function via trace amine-associated receptor 1: implication for modulatory roles of trace amines in brain. J Pharmacol Exp Ther. 2008;325:617–28.CrossRefPubMed Xie Z, Miller GM. Beta-phenylethylamine alters monoamine transporter function via trace amine-associated receptor 1: implication for modulatory roles of trace amines in brain. J Pharmacol Exp Ther. 2008;325:617–28.CrossRefPubMed
9.
go back to reference Bradaia A, Trube G, Stalder H, Norcross RD, Ozmen L, Wettstein JG, et al. The selective antagonist EPPTB reveals TAAR1-mediated regulatory mechanisms in dopaminergic neurons of the mesolimbic system. Proc Natl Acad Sci USA. 2009;106:20081–6.CrossRefPubMedPubMedCentral Bradaia A, Trube G, Stalder H, Norcross RD, Ozmen L, Wettstein JG, et al. The selective antagonist EPPTB reveals TAAR1-mediated regulatory mechanisms in dopaminergic neurons of the mesolimbic system. Proc Natl Acad Sci USA. 2009;106:20081–6.CrossRefPubMedPubMedCentral
10.
go back to reference Leo D, Mus L, Espinoza S, Hoener MC, Sotnikova TD, Gainetdinov RR. Taar1-mediated modulation of presynaptic dopaminergic neurotransmission: role of D2 dopamine autoreceptors. Neuropharmacology. 2014;81:283–91.CrossRefPubMed Leo D, Mus L, Espinoza S, Hoener MC, Sotnikova TD, Gainetdinov RR. Taar1-mediated modulation of presynaptic dopaminergic neurotransmission: role of D2 dopamine autoreceptors. Neuropharmacology. 2014;81:283–91.CrossRefPubMed
11.
go back to reference Oldendorf WH. Brain uptake of radiolabeled amino acids, amines, and hexoses after arterial injection. Am J Physiol. 1971;221:1629–39.PubMed Oldendorf WH. Brain uptake of radiolabeled amino acids, amines, and hexoses after arterial injection. Am J Physiol. 1971;221:1629–39.PubMed
12.
go back to reference James JH, Escourrou J, Fischer JE. Blood-brain neutral amino acid transport activity is increased after portacaval anastomosis. Science. 1978;200:1395–7.CrossRefPubMed James JH, Escourrou J, Fischer JE. Blood-brain neutral amino acid transport activity is increased after portacaval anastomosis. Science. 1978;200:1395–7.CrossRefPubMed
13.
go back to reference Faraj BA, Camp VM, Ansley JD, Scott J, Ali FM, Malveaux EJ. Evidence for central hypertyraminemia in hepatic encephalopathy. J Clin Invest. 1981;67:395–402.CrossRefPubMedPubMedCentral Faraj BA, Camp VM, Ansley JD, Scott J, Ali FM, Malveaux EJ. Evidence for central hypertyraminemia in hepatic encephalopathy. J Clin Invest. 1981;67:395–402.CrossRefPubMedPubMedCentral
14.
go back to reference Berry MD. Mammalian central nervous system trace amines. Pharmacologic amphetamines, physiologic neuromodulators. J Neurochem. 2004;90:257–71.CrossRefPubMed Berry MD. Mammalian central nervous system trace amines. Pharmacologic amphetamines, physiologic neuromodulators. J Neurochem. 2004;90:257–71.CrossRefPubMed
15.
go back to reference Ohtsuki S, Terasaki T. Contribution of carrier-mediated transport systems to the blood–brain barrier as a supporting and protecting interface for the brain; importance for CNS drug discovery and development. Pharm Res. 2007;24:1745–58.CrossRefPubMed Ohtsuki S, Terasaki T. Contribution of carrier-mediated transport systems to the blood–brain barrier as a supporting and protecting interface for the brain; importance for CNS drug discovery and development. Pharm Res. 2007;24:1745–58.CrossRefPubMed
16.
go back to reference Usui T, Nakazawa A, Okura T, Deguchi Y, Akanuma SI, Kubo Y, et al. Histamine elimination from the cerebrospinal fluid across the blood-cerebrospinal fluid barrier: involvement of plasma membrane monoamine transporter (PMAT/SLC29A4). J Neurochem. 2016;139:408–18.CrossRefPubMed Usui T, Nakazawa A, Okura T, Deguchi Y, Akanuma SI, Kubo Y, et al. Histamine elimination from the cerebrospinal fluid across the blood-cerebrospinal fluid barrier: involvement of plasma membrane monoamine transporter (PMAT/SLC29A4). J Neurochem. 2016;139:408–18.CrossRefPubMed
17.
go back to reference Mori S, Takanaga H, Ohtsuki S, Deguchi T, Kang YS, Hosoya K, et al. Rat organic anion transporter 3 (rOAT3) is responsible for brain-to-blood efflux of homovanillic acid at the abluminal membrane of brain capillary endothelial cells. J Cereb Blood Flow Metab. 2003;23:432–40.CrossRefPubMed Mori S, Takanaga H, Ohtsuki S, Deguchi T, Kang YS, Hosoya K, et al. Rat organic anion transporter 3 (rOAT3) is responsible for brain-to-blood efflux of homovanillic acid at the abluminal membrane of brain capillary endothelial cells. J Cereb Blood Flow Metab. 2003;23:432–40.CrossRefPubMed
18.
go back to reference Okura T, Kato S, Takano Y, Sato T, Yamashita A, Morimoto R, et al. Functional characterization of rat plasma membrane monoamine transporter in the blood–brain and blood-cerebrospinal fluid barriers. J Pharm Sci. 2011;100:3924–38.CrossRefPubMed Okura T, Kato S, Takano Y, Sato T, Yamashita A, Morimoto R, et al. Functional characterization of rat plasma membrane monoamine transporter in the blood–brain and blood-cerebrospinal fluid barriers. J Pharm Sci. 2011;100:3924–38.CrossRefPubMed
19.
go back to reference Wakayama K, Ohtsuki S, Takanaga H, Hosoya K, Terasaki T. Localization of norepinephrine and serotonin transporter in mouse brain capillary endothelial cells. Neurosci Res. 2002;44:173–80.CrossRefPubMed Wakayama K, Ohtsuki S, Takanaga H, Hosoya K, Terasaki T. Localization of norepinephrine and serotonin transporter in mouse brain capillary endothelial cells. Neurosci Res. 2002;44:173–80.CrossRefPubMed
20.
go back to reference Tachikawa M, Kasai Y, Takahashi M, Fujinawa J, Kitaichi K, Terasaki T, et al. The blood-cerebrospinal fluid barrier is a major pathway of cerebral creatinine clearance: involvement of transporter-mediated process. J Neurochem. 2008;107:432–42.CrossRefPubMed Tachikawa M, Kasai Y, Takahashi M, Fujinawa J, Kitaichi K, Terasaki T, et al. The blood-cerebrospinal fluid barrier is a major pathway of cerebral creatinine clearance: involvement of transporter-mediated process. J Neurochem. 2008;107:432–42.CrossRefPubMed
21.
go back to reference Chen L, Pawlikowski B, Schlessinger A, More SS, Stryke D, Johns SJ, et al. Role of organic cation transporter 3 (SLC22A3) and its missense variants in the pharmacologic action of metformin. Pharmacogenet Genom. 2010;20:687–99.CrossRef Chen L, Pawlikowski B, Schlessinger A, More SS, Stryke D, Johns SJ, et al. Role of organic cation transporter 3 (SLC22A3) and its missense variants in the pharmacologic action of metformin. Pharmacogenet Genom. 2010;20:687–99.CrossRef
22.
go back to reference Engel K, Wang J. Interaction of organic cations with a newly identified plasma membrane monoamine transporter. Mol Pharmacol. 2005;68:1397–407.CrossRefPubMed Engel K, Wang J. Interaction of organic cations with a newly identified plasma membrane monoamine transporter. Mol Pharmacol. 2005;68:1397–407.CrossRefPubMed
23.
go back to reference Hilber B, Scholze P, Dorostkar MM, Sandtner W, Holy M, Boehm S, et al. Serotonin-transporter mediated efflux: a pharmacological analysis of amphetamines and non-amphetamines. Neuropharmacology. 2005;49:811–9.CrossRefPubMed Hilber B, Scholze P, Dorostkar MM, Sandtner W, Holy M, Boehm S, et al. Serotonin-transporter mediated efflux: a pharmacological analysis of amphetamines and non-amphetamines. Neuropharmacology. 2005;49:811–9.CrossRefPubMed
24.
go back to reference Sitte HH, Huck S, Reither H, Boehm S, Singer EA, Pifl C. Carrier-mediated release, transport rates, and charge transfer induced by amphetamine, tyramine, and dopamine in mammalian cells transfected with the human dopamine transporter. J Neurochem. 1998;71:1289–97.CrossRefPubMed Sitte HH, Huck S, Reither H, Boehm S, Singer EA, Pifl C. Carrier-mediated release, transport rates, and charge transfer induced by amphetamine, tyramine, and dopamine in mammalian cells transfected with the human dopamine transporter. J Neurochem. 1998;71:1289–97.CrossRefPubMed
25.
go back to reference Kakee A, Terasaki T, Sugiyama Y. Brain efflux index as a novel method of analyzing efflux transport at the blood–brain barrier. J Pharmacol Exp Ther. 1996;277:1550–9.PubMed Kakee A, Terasaki T, Sugiyama Y. Brain efflux index as a novel method of analyzing efflux transport at the blood–brain barrier. J Pharmacol Exp Ther. 1996;277:1550–9.PubMed
26.
go back to reference Ogawa M, Suzuki H, Sawada Y, Hanano M, Sugiyama Y. Kinetics of active efflux via choroid plexus of beta-lactam antibiotics from the CSF into the circulation. Am J Physiol. 1994;266:R392–9.PubMed Ogawa M, Suzuki H, Sawada Y, Hanano M, Sugiyama Y. Kinetics of active efflux via choroid plexus of beta-lactam antibiotics from the CSF into the circulation. Am J Physiol. 1994;266:R392–9.PubMed
27.
go back to reference Suzuki H, Sawada Y, Sugiyama Y, Iga T, Hanano M. Transport of benzylpenicillin by the rat choroid plexus in vitro. J Pharmacol Exp Ther. 1987;242:660–5.PubMed Suzuki H, Sawada Y, Sugiyama Y, Iga T, Hanano M. Transport of benzylpenicillin by the rat choroid plexus in vitro. J Pharmacol Exp Ther. 1987;242:660–5.PubMed
28.
go back to reference Suzuki H, Sawada Y, Sugiyama Y, Iga T, Hanano M. Transport of cimetidine by the rat choroid plexus in vitro. J Pharmacol Exp Ther. 1986;239:927–35.PubMed Suzuki H, Sawada Y, Sugiyama Y, Iga T, Hanano M. Transport of cimetidine by the rat choroid plexus in vitro. J Pharmacol Exp Ther. 1986;239:927–35.PubMed
29.
go back to reference Kitazawa T, Hosoya K, Watanabe M, Takashima T, Ohtsuki S, Takanaga H, et al. Characterization of the amino acid transport of new immortalized choroid plexus epithelial cell lines: a novel in vitro system for investigating transport functions at the blood-cerebrospinal fluid barrier. Pharm Res. 2001;18:16–22.CrossRefPubMed Kitazawa T, Hosoya K, Watanabe M, Takashima T, Ohtsuki S, Takanaga H, et al. Characterization of the amino acid transport of new immortalized choroid plexus epithelial cell lines: a novel in vitro system for investigating transport functions at the blood-cerebrospinal fluid barrier. Pharm Res. 2001;18:16–22.CrossRefPubMed
30.
go back to reference Yamaoka K, Tanigawara Y, Nakagawa T, Uno T. A pharmacokinetic analysis program (multi) for microcomputer. J Pharmacobiodyn. 1981;4:879–85.CrossRefPubMed Yamaoka K, Tanigawara Y, Nakagawa T, Uno T. A pharmacokinetic analysis program (multi) for microcomputer. J Pharmacobiodyn. 1981;4:879–85.CrossRefPubMed
31.
go back to reference Ohta KY, Inoue K, Hayashi Y, Yuasa H. Molecular identification and functional characterization of rat multidrug and toxin extrusion type transporter 1 as an organic cation/H+ antiporter in the kidney. Drug Metab Dispos. 2006;34:1868–74.CrossRefPubMed Ohta KY, Inoue K, Hayashi Y, Yuasa H. Molecular identification and functional characterization of rat multidrug and toxin extrusion type transporter 1 as an organic cation/H+ antiporter in the kidney. Drug Metab Dispos. 2006;34:1868–74.CrossRefPubMed
32.
go back to reference Suzuki H, Sawada Y, Sugiyama Y, Iga T, Hanano M. Saturable transport of cimetidine from cerebrospinal fluid to blood in rats. J Pharmacobiodyn. 1985;8:73–6.CrossRefPubMed Suzuki H, Sawada Y, Sugiyama Y, Iga T, Hanano M. Saturable transport of cimetidine from cerebrospinal fluid to blood in rats. J Pharmacobiodyn. 1985;8:73–6.CrossRefPubMed
33.
go back to reference Akanuma S, Sakurai T, Tachikawa M, Kubo Y, Hosoya K. Transporter-mediated l-glutamate elimination from cerebrospinal fluid: possible involvement of excitatory amino acid transporters expressed in ependymal cells and choroid plexus epithelial cells. Fluids Barriers CNS. 2015;12:11.CrossRefPubMedPubMedCentral Akanuma S, Sakurai T, Tachikawa M, Kubo Y, Hosoya K. Transporter-mediated l-glutamate elimination from cerebrospinal fluid: possible involvement of excitatory amino acid transporters expressed in ependymal cells and choroid plexus epithelial cells. Fluids Barriers CNS. 2015;12:11.CrossRefPubMedPubMedCentral
34.
go back to reference Quay WB. Regional differences in metabolism and composition of choroid plexuses. Brain Res. 1966;2:378–89.CrossRefPubMed Quay WB. Regional differences in metabolism and composition of choroid plexuses. Brain Res. 1966;2:378–89.CrossRefPubMed
35.
go back to reference Krunic N, Adamson SL, Coceani F. Differential uptake and catabolism of prostaglandin (PG)E(2) versus PGF(2alpha) in the sheep choroid plexus during development. Brain Res Dev Brain Res. 2000;119:11–9.CrossRefPubMed Krunic N, Adamson SL, Coceani F. Differential uptake and catabolism of prostaglandin (PG)E(2) versus PGF(2alpha) in the sheep choroid plexus during development. Brain Res Dev Brain Res. 2000;119:11–9.CrossRefPubMed
36.
go back to reference Keep RF, Jones HC. A morphometric study on the development of the lateral ventricle choroid plexus, choroid plexus capillaries and ventricular ependyma in the rat. Brain Res Dev Brain Res. 1990;56:47–53.CrossRefPubMed Keep RF, Jones HC. A morphometric study on the development of the lateral ventricle choroid plexus, choroid plexus capillaries and ventricular ependyma in the rat. Brain Res Dev Brain Res. 1990;56:47–53.CrossRefPubMed
37.
go back to reference Fukao M, Ishida K, Horie A, Taguchi M, Nozawa T, Inoue H, et al. Variability of bioavailability and intestinal absorption mechanisms of metoprolol. Drug Metab Pharmacokinet. 2014;29:162–7.CrossRefPubMed Fukao M, Ishida K, Horie A, Taguchi M, Nozawa T, Inoue H, et al. Variability of bioavailability and intestinal absorption mechanisms of metoprolol. Drug Metab Pharmacokinet. 2014;29:162–7.CrossRefPubMed
38.
go back to reference Ishida K, Horie A, Nishimura M, Taguchi M, Fujii N, Nozawa T, et al. Variability of bioavailability and intestinal absorption characteristics of bisoprolol. Drug Metab Pharmacokinet. 2013;28:491–6.CrossRefPubMed Ishida K, Horie A, Nishimura M, Taguchi M, Fujii N, Nozawa T, et al. Variability of bioavailability and intestinal absorption characteristics of bisoprolol. Drug Metab Pharmacokinet. 2013;28:491–6.CrossRefPubMed
39.
go back to reference Tega Y, Akanuma S, Kubo Y, Hosoya K. Involvement of the H+/organic cation antiporter in nicotine transport in rat liver. Drug Metab Dispos. 2015;43:89–92.CrossRefPubMed Tega Y, Akanuma S, Kubo Y, Hosoya K. Involvement of the H+/organic cation antiporter in nicotine transport in rat liver. Drug Metab Dispos. 2015;43:89–92.CrossRefPubMed
40.
go back to reference Tega Y, Yuzurihara C, Kubo Y, Akanuma SI, Ehrhardt C, Hosoya KI. Functional expression of nicotine influx transporter in A549 human alveolar epithelial cells. Drug Metab Pharmacokinet. 2016;31:99–101.CrossRefPubMed Tega Y, Yuzurihara C, Kubo Y, Akanuma SI, Ehrhardt C, Hosoya KI. Functional expression of nicotine influx transporter in A549 human alveolar epithelial cells. Drug Metab Pharmacokinet. 2016;31:99–101.CrossRefPubMed
41.
go back to reference Tega Y, Akanuma S, Kubo Y, Terasaki T, Hosoya K. Blood-to-brain influx transport of nicotine at the rat blood–brain barrier: involvement of a pyrilamine-sensitive organic cation transport process. Neurochem Int. 2013;62:173–81.CrossRefPubMed Tega Y, Akanuma S, Kubo Y, Terasaki T, Hosoya K. Blood-to-brain influx transport of nicotine at the rat blood–brain barrier: involvement of a pyrilamine-sensitive organic cation transport process. Neurochem Int. 2013;62:173–81.CrossRefPubMed
42.
go back to reference Okura T, Hattori A, Takano Y, Sato T, Hammarlund-Udenaes M, Terasaki T, et al. Involvement of the pyrilamine transporter, a putative organic cation transporter, in blood–brain barrier transport of oxycodone. Drug Metab Dispos. 2008;36:2005–13.CrossRefPubMed Okura T, Hattori A, Takano Y, Sato T, Hammarlund-Udenaes M, Terasaki T, et al. Involvement of the pyrilamine transporter, a putative organic cation transporter, in blood–brain barrier transport of oxycodone. Drug Metab Dispos. 2008;36:2005–13.CrossRefPubMed
43.
go back to reference Yamazaki M, Fukuoka H, Nagata O, Kato H, Ito Y, Terasaki T, et al. Transport mechanism of an H1-antagonist at the blood–brain barrier: transport mechanism of mepyramine using the carotid injection technique. Biol Pharm Bull. 1994;17:676–9.CrossRefPubMed Yamazaki M, Fukuoka H, Nagata O, Kato H, Ito Y, Terasaki T, et al. Transport mechanism of an H1-antagonist at the blood–brain barrier: transport mechanism of mepyramine using the carotid injection technique. Biol Pharm Bull. 1994;17:676–9.CrossRefPubMed
44.
go back to reference Youdim MB, Bakhle YS. Monoamine oxidase: isoforms and inhibitors in Parkinson’s disease and depressive illness. Br J Pharmacol. 2006;147(Suppl 1):S287–96.PubMedPubMedCentral Youdim MB, Bakhle YS. Monoamine oxidase: isoforms and inhibitors in Parkinson’s disease and depressive illness. Br J Pharmacol. 2006;147(Suppl 1):S287–96.PubMedPubMedCentral
Metadata
Title
Role of cationic drug-sensitive transport systems at the blood-cerebrospinal fluid barrier in para-tyramine elimination from rat brain
Authors
Shin-ichi Akanuma
Yuhei Yamazaki
Yoshiyuki Kubo
Ken-ichi Hosoya
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Fluids and Barriers of the CNS / Issue 1/2018
Electronic ISSN: 2045-8118
DOI
https://doi.org/10.1186/s12987-017-0087-9

Other articles of this Issue 1/2018

Fluids and Barriers of the CNS 1/2018 Go to the issue