Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters











Publication year range
1.
J Pharm Sci ; 112(12): 3216-3223, 2023 12.
Article in English | MEDLINE | ID: mdl-37690777

ABSTRACT

In order to establish an in vitro model of the human blood-brain barrier (BBB), MDR1-overexpressing human induced pluripotent stem cells (hiPSCs) were generated, and they were differentiated to MDR1-expressing brain microvascular endothelial-like cells (MDR1-expressing hiPS-BMECs). MDR1-expressing hiPS-BMECs monolayers showed good barrier function in terms of tight junction protein expression and trans-epithelial electrical resistance (TEER). In sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH-MS), MDR1 protein expression was markedly increased in MDR1-expressing hiPS-BMECs, whereas other ABC and SLC transporters showed almost identical expression between MDR1-expressing hiPS-BMECs and mock hiPS-BMECs, suggesting that MDR1 overexpression had little or no knock-on effect on other proteins. The basolateral-to-apical transport of MDR1 substrates, such as quinidine, [3H]digoxin and [3H]vinblastine, was higher than the apical-to-basolateral transport, and the efflux-dominant transport was attenuated by PSC833, an MDR1-specific inhibitor, indicating that MDR1-mediated efflux transport is functional. The robust MDR1 function was also supported by the efflux-dominant transports of [3H]cyclosporin A, loperamide, cetirizine, and verapamil by MDR1-expressing hiPS-BMECs. These results suggest that MDR1-expressing hiPS-BMECs can be used as an in vitro model of the human BBB.


Subject(s)
Blood-Brain Barrier , Induced Pluripotent Stem Cells , Humans , Brain , Cell Line , Cells, Cultured
2.
Biol Pharm Bull ; 45(10): 1585-1589, 2022.
Article in English | MEDLINE | ID: mdl-36184519

ABSTRACT

Naltrexone is a mu-opioid receptor antagonist used in the treatment of opioid and alcohol dependence. The blood-brain barrier (BBB) transport characteristics of naltrexone was investigated by means of hCMEC/D3 cells, a human immortalized brain capillary endothelial cell line. In hCMEC/D3 cells, naltrexone is taken up in a concentration-dependent manner. Furthermore, naltrexone uptake significantly decreased in the presence of H+/organic cation (OC) antiporter substrates, during the little alteration exhibited by substrates of well-identified OC transporters classified into SLC22A family. Although naltrexone uptake by hCMEC/D3 cells was partially affected by changes of ionic conditions, it was markedly decreased in the presence of the metabolic inhibitor sodium azide. Furthermore, when treated by ammonium chloride, naltrexone uptake by hCMEC/D3 cells was altered by intracellular acidification and alkalization, suggesting the involvement of oppositely directed proton gradient in naltrexone transport across the BBB. The results obtained in the present in vitro study suggest the active transport of naltrexone from blood to the brain across the BBB by the H+/OC antiporter.


Subject(s)
Antiporters , Blood-Brain Barrier , Ammonium Chloride , Analgesics, Opioid/metabolism , Antiporters/metabolism , Biological Transport , Blood-Brain Barrier/metabolism , Cations/metabolism , Humans , Naltrexone/metabolism , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Protons , Sodium Azide/metabolism
3.
Pharmaceutics ; 14(8)2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36015309

ABSTRACT

A proton-coupled organic cation (H+/OC) antiporter working at the blood-brain barrier (BBB) in humans and rodents is thought to be a promising candidate for the efficient delivery of cationic drugs to the brain. Therefore, it is important to identify the molecular entity that exhibits this activity. Here, for this purpose, we established the Proteomics-based Identification of transporter by Crosslinking substrate in Keyhole (PICK) method, which combines photo-affinity labeling with comprehensive proteomics analysis using SWATH-MS. Using preselected criteria, the PICK method generated sixteen candidate proteins. From these, knockdown screening in hCMEC/D3 cells, an in vitro BBB model, identified two proteins, TM7SF3 and LHFPL6, as candidates for the H+/OC antiporter. We synthesized a novel H+/OC antiporter substrate for functional analysis of TM7SF3 and LHFPL6 in hCMEC/D3 cells and HEK293 cells. The results suggested that both TM7SF3 and LHFPL6 are components of the H+/OC antiporter.

6.
Pharm Res ; 39(7): 1535-1547, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35411503

ABSTRACT

PURPOSE: The purpose of this study was to construct and validate an in vitro three-dimensional blood-brain barrier (3DBBB) model system equipped with brain microvascular endothelial cells derived from human induced pluripotent stem cells (hiPS-BMECs). METHODS: The 3D-BBB system was constructed by seeding hiPS-BMECs onto the capillary lane of a MIMETAS OrganoPlate® 3-lane coated with fibronectin/collagen IV. hiPS-BMECs were incubated under continuous switchback flow with an OrganoFlow® for 2 days. The 3D capillary structure and expression of tight-junction proteins and transporters were confirmed by immunocytochemistry. The mRNA expression of transporters in the 3D environment was determined using qRT-PCR, and the permeability of endogenous substances and drugs was evaluated under various conditions. RESULTS AND DISCUSSION: The expression of tight-junction proteins, including claudin-5 and ZO-1, was confirmed by immunohistochemistry. The permeability rate constant of lucifer yellow through hiPS-BMECs was undetectably low, indicating that paracellular transport is highly restricted by tight junctions in the 3D-BBB system. The mRNA expression levels of transporters and receptors in the 3D-BBB system differed from those in the 2D-culture system by 0.2- to 5.8-fold. The 3D-cultured hiPS-BMECs showed asymmetric transport of substrates of BCRP, CAT1 and LAT1 between the luminal (blood) and abluminal (brain) sides. Proton-coupled symport function of MCT1 was also confirmed. CONCLUSION: The 3D-BBB system constructed in this study mimics several important characteristics of the human BBB, and is expected to be a useful high-throughput evaluation tool in the development of CNS drugs.


Subject(s)
Blood-Brain Barrier , Brain , Endothelial Cells , Induced Pluripotent Stem Cells , Blood-Brain Barrier/metabolism , Brain/blood supply , Cells, Cultured , Endothelial Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Membrane Transport Proteins/metabolism , RNA, Messenger/metabolism , Tight Junction Proteins/metabolism
7.
Pharm Res ; 39(7): 1549-1559, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35314999

ABSTRACT

AIM: Identification of blood-brain barrier (BBB) uptake transporters is a major challenge in the research and development of central nervous system (CNS) drugs. However, conventional methods that consider known drug uptake characteristics have failed at identifying the responsible transporter molecule. The present study aimed at identifying aripiprazole uptake transporters in BBB model hCMEC/D3 cells using a knockdown screening study targeting various transporters, including uncharacterized ones. METHODS: We evaluated the effect of 214 types of siRNA targeting transporters on the uptake of aripiprazole, an atypical antipsychotic drug, in hCMEC/D3 cells. Aripiprazole uptake was determined using Xenopus oocytes expressing the candidate genes extracted from the siRNA screening assay. RESULTS: The estimated unbound brain to plasma concentration ratio (Kp,uu,brain) of aripiprazole was estimated as 0.67 in wild-type mice and 1.94 in abcb1a/1b/abcg2 knockout mice, suggesting the involvement of both uptake and efflux transporters in BBB permeation. According to siRNA knockdown screening studies, organic cation/carnitine transporter 2 (OCTN2) and long-chain fatty acid transporter 1 (FATP1) were identified as candidate genes. The uptake of aripiprazole by hCMEC/D3 cells was decreased by OCTN2 inhibitors, but not by FATP1 inhibitors. A partially increased uptake of aripiprazole was observed in OCTN2-expressing Xenopus oocytes. Finally, to evaluate transporter-mediated BBB permeation of drugs, the reported and estimated Kp,uu,brain values were summarized. CONCLUSIONS: A knockdown screening study in combination with Kp,uu,brain values showed that aripiprazole was a potential substrate of OCTN2. The technique described in this study can be applied to identifying novel BBB transporters for CNS drugs.


Subject(s)
Blood-Brain Barrier , Membrane Transport Proteins , Animals , Aripiprazole/pharmacology , Biological Transport , Brain , Mice , RNA, Small Interfering/genetics
8.
J Pharm Sci ; 110(10): 3484-3490, 2021 10.
Article in English | MEDLINE | ID: mdl-34102205

ABSTRACT

The likelihood of reoccurrence of acute lymphoblastic leukemia is influenced by the cerebral concentration of the therapeutic agent 6-mercaptopurine (6-MP) during treatment. Therefore, it is important to understand the blood-brain barrier (BBB) transport mechanism of 6-MP. The purpose of this study was to characterize this mechanism using human induced pluripotent stem cell-derived microvascular endothelial cells (hiPS-BMECs). The permeability coefficient of 6-MP across hiPS-BMECs monolayer in the basal-to-apical direction (B-to-A) was significantly greater than that in the opposite direction (A-to-B). The inhibition profiles of 6-MP transport in the A-to-B direction were different from those in the B-to-A direction. Transport in the A-to-B direction was mainly inhibited by adenine (an inhibitor of equilibrative nucleobase transporter 1; ENBT1), while transport in the B-to-A direction was significantly reduced by inhibitors of multidrug resistance-associated proteins (MRPs), especially zaprinast (an MRP5 inhibitor). Immunocytochemical analyses demonstrated the expression of ENBT1 and MRP5 proteins in hiPS-BMECs. We confirmed that the cellular uptake of 6-MP is decreased by ENBT1 inhibitors in hiPS-BMECs and by knockdown of ENBT1 in hCMEC/D3 cells. These results suggest that ENBT1 and MRP5 make substantial contributions to the transport of 6-MP in hiPS-BMECs and hCMEC/D3 cells.


Subject(s)
Induced Pluripotent Stem Cells , Mercaptopurine , Biological Transport , Blood-Brain Barrier , Brain , Endothelial Cells , Humans
9.
Drug Metab Dispos ; 49(1): 3-11, 2021 01.
Article in English | MEDLINE | ID: mdl-33144341

ABSTRACT

Understanding the mechanisms of drug transport across the blood-brain barrier (BBB) is an important issue for regulating the pharmacokinetics of drugs in the central nervous system. In this study, we focused on solute carrier family 35, member F2 (SLC35F2), whose mRNA is highly expressed in the BBB. SLC35F2 protein was enriched in isolated mouse and monkey brain capillaries relative to brain homogenates and was localized exclusively on the apical membrane of MDCKII cells and brain microvascular endothelial cells (BMECs) differentiated from human induced pluripotent stem cells (hiPS-BMECs). SLC35F2 activity was assessed using its substrate, YM155, and pharmacological experiments revealed SLC35F2 inhibitors, such as famotidine (half-maximal inhibitory concentration, 160 µM). Uptake of YM155 was decreased by famotidine or SLC35F2 knockdown in immortalized human BMECs (human cerebral microvascular endothelial cell/D3 cells). Furthermore, famotidine significantly inhibited the apical (A)-to-basal (B) transport of YM155 in primary cultured monkey BMECs and hiPS-BMECs. Crucially, SLC35F2 knockout diminished the A-to-B transport and intracellular accumulation of YM155 in hiPS-BMECs. By contrast, in studies using an in situ brain perfusion technique, neither deletion of Slc35f2 nor famotidine reduced brain uptake of YM155, even though YM155 is a substrate of mouse SLC35F2. YM155 uptake was decreased significantly by losartan and naringin, inhibitors for the organic anion transporting polypeptide (OATP) 1A4. These findings suggest SLC35F2 is a functional transporter in various cellular models of the primate BBB that delivers its substrates to the brain and that its relative importance in the BBB is modified by differences in the expression of OATPs between primates and rodents. SIGNIFICANCE STATEMENT: This study demonstrated that SLC35F2 is a functional drug influx transporter in three different cellular models of the primate blood-brain barrier (i.e., human cerebral microvascular endothelial cell/D3 cells, primary cultured monkey BMECs, and human induced pluripotent stem-BMECs) but has limited roles in mouse brain. SLC35F2 facilitates apical-to-basal transport across the tight cell monolayer. These findings will contribute to the development of improved strategies for targeting drugs to the central nervous system.


Subject(s)
Biological Transport/drug effects , Blood-Brain Barrier , Famotidine/pharmacokinetics , Imidazoles/pharmacokinetics , Membrane Transport Proteins/metabolism , Naphthoquinones/pharmacokinetics , Organic Anion Transporters/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cells, Cultured , Central Nervous System Agents/pharmacokinetics , Drug Development/methods , Endothelial Cells/metabolism , Haplorhini , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Models, Biological
10.
Cell Tissue Res ; 383(2): 823-833, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32910242

ABSTRACT

Endothelial fenestrae are transcellular pores that pierce the capillary walls in endocrine glands such as the pituitary. The fenestrae are covered with a thin fibrous diaphragm consisting of the plasmalemma vesicle-associated protein (PLVAP) that clusters to form sieve plates. The basal surface of the vascular wall is lined by basement membrane (BM) composed of various extracellular matrices (ECMs). However, the relationship between the ECMs and the endothelial fenestrae is still unknown. In this study, we isolated fenestrated endothelial cells from the anterior lobe of the rat pituitary, using a dynabeads-labeled antibody against platelet endothelial cell adhesion molecule 1 (PECAM1). We then analyzed the gene expression levels of several endothelial marker genes and genes for integrin α subunits, which function as the receptors for ECMs, by real-time polymerase chain reaction (PCR). The results showed that the genes for the integrin α subunit, which binds to collagen IV, fibronectin, laminin-411, or laminin-511, were highly expressed. When the PECAM1-positive cells were cultured for 7 days on collagen IV-, fibronectin-, laminins-411-, or laminins-511-coated coverslips, the sieve plate structures equipped with probably functional fenestrae were maintained only when the cells were cultured on fibronectin. Additionally, real-time PCR analysis showed that the fibronectin coating was effective in maintaining the expression pattern of several endothelial marker genes that were preferentially expressed in the endothelial cells of the fenestrated capillaries. These results indicate that fibronectin functions as the principal factor in the maintenance of the sieve plate structures in the endothelial cells of the fenestrated capillary.


Subject(s)
Capillaries/metabolism , Endothelial Cells/metabolism , Fibronectins/metabolism , Animals , Biomarkers/metabolism , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Endothelial Cells/ultrastructure , Male , Membrane Proteins/metabolism , Pituitary Gland/cytology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rats, Wistar
11.
J Pharm Sci ; 110(1): 397-403, 2021 01.
Article in English | MEDLINE | ID: mdl-32898521

ABSTRACT

There is increasing evidence that a proton-coupled organic cation (H+/OC) antiporter facilitates uptake of various central nervous system-active drugs, such as the histamine H1 receptor antagonist diphenhydramine, into the brain. The purpose of this study was to clarify the structural requirements for H+/OC antiporter-mediated uptake into hCMEC/D3 cells, an established in vitro model of the human blood-brain barrier, by using a series of diphenhydramine analogs. For this purpose, we synthesized seven tertiary amine analogs and three amide analogs. Uptake of all the amines was facilitated by an outwardly directed H+ gradient and inhibited by pyrilamine, a typical substrate and a strong inhibitor of the H+/OC antiporter. Further, uptake of most of the amines was trans-stimulated by pyrilamine. Uptake of the amines was 21 times faster than that of the amides on average, even though the lipophilicity (log D7.4) of the amines is lower than that of the amides. Amines containing a pyrrolidine or piperidine ring showed the highest uptake rates. Our results suggest that an amine moiety, especially a heterocyclic amine moiety, is important for recognition and transport by the H+/OC antiporter.


Subject(s)
Antiporters , Protons , Antiporters/metabolism , Biological Transport , Blood-Brain Barrier/metabolism , Cations , Diphenhydramine , Humans , Organic Cation Transport Proteins/metabolism
12.
Mol Pharm ; 15(12): 5546-5555, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30376629

ABSTRACT

Brain microvascular endothelial cells derived from human induced pluripotent stem cells (hiPS-BMECs) have been proposed as a new blood-brain barrier model, but their transport function has not been fully clarified. Therefore, in this study, we investigated the gene expression and function of transporters in hiPS-BMECs by means of quantitative reverse transcription-PCR, in vitro transcellular transport studies, and uptake experiments. mRNAs encoding ABC and SLC transporters, such as BCRP, MCT1, CAT1, and GLAST, were highly expressed in hiPS-BMECs. Transcellular transport studies showed that prazosin, [14C]l-lactate, [3H]l-arginine, and [3H]l-glutamate (substrates of BCRP, MCT1, CAT1, and GLAST, respectively) were transported asymmetrically across the hiPS-BMEC monolayer. Substrates of LAT1, OCTN2, CAT1, GLAST, MCT1, and proton-coupled organic cation (H+/OC) antiporter were taken up by hiPS-BMECs in a time-, temperature-, and concentration-dependent manner, and the uptakes were markedly decreased by inhibitors of the corresponding transporter. These results indicate that hiPS-BMECs express multiple nutrient and drug transporters.


Subject(s)
Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Membrane Transport Proteins/metabolism , Arginine/pharmacokinetics , Cell Differentiation , Cell Line , Cell Membrane Permeability/drug effects , Glutamic Acid/pharmacokinetics , Humans , Induced Pluripotent Stem Cells/physiology , Lactic Acid/pharmacokinetics , Membrane Transport Proteins/genetics , Microvessels/cytology , Prazosin/pharmacokinetics , RNA, Messenger/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
13.
Pharm Res ; 35(12): 246, 2018 Oct 29.
Article in English | MEDLINE | ID: mdl-30374619

ABSTRACT

PURPOSE: The anti-epileptic drug pregabalin crosses the blood-brain barrier (BBB) in spite of its low lipophilicity. This study was performed to determine whether L-type amino acid transporters (LAT1/SLC7A5 and LAT2/SLC7A8) contribute to the uptake of pregabalin. METHODS: Pregabalin uptake by LATs-transfected HEK293 cells or hCMEC/D3 cells, an in vitro human BBB model, was measured by LC-MS/MS analysis. Expression of LAT1 mRNA in hCMEC/D3 cells was determined by quantitative RT-PCR analysis. RESULTS: Overexpression of LAT1, but not LAT2, in HEK293 cells significantly increased the cellular uptake of pregabalin, and the LAT1-mediated uptake was saturable with a Km of 0.288 mM. LAT1-mediated amino acid uptake was inhibited specifically and almost completely in the presence of 1 mM pregabalin. The uptake of pregabalin by hCMEC/D3 cells was sodium-independent, saturable (Km = 0.854 mM), and strongly inhibited by large amino acids at 1 mM, 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid, a specific system L inhibitor, at 1 mM and by JPH203, a LAT1-selective inhibitor, at 10 µM. Pregabalin uptake in hCMEC/D3 cells was also decreased by 75% by the silencing of LAT1 gene using LAT1 siRNA. CONCLUSIONS: Our results indicate that LAT1, but not LAT2, recognizes pregabalin as a substrate. It is suggested that LAT1 mediates pregabalin transport at the BBB.


Subject(s)
Anticonvulsants/pharmacokinetics , Blood-Brain Barrier/metabolism , Endothelial Cells/drug effects , Large Neutral Amino Acid-Transporter 1/metabolism , Pregabalin/pharmacokinetics , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Animals , Anticonvulsants/metabolism , Biological Transport , Brain/blood supply , Cell Line , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Fusion Regulatory Protein 1, Light Chains/genetics , Fusion Regulatory Protein 1, Light Chains/metabolism , Humans , Large Neutral Amino Acid-Transporter 1/genetics , Leucine/metabolism , Permeability , Pregabalin/metabolism , RNA, Small Interfering/genetics , Rats
14.
ACS Med Chem Lett ; 9(9): 884-888, 2018 Sep 13.
Article in English | MEDLINE | ID: mdl-30258535

ABSTRACT

We designed and synthesized a pyrilamine derivative 1 as a selective class I HDAC inhibitor that targets pyrilamine-sensitive proton-coupled organic cation antiporter (PYSOCA) at the blood-brain barrier (BBB). Introduction of pyrilamine moiety to benzamide type HDAC inhibitors kept selective class I HDAC inhibitory activity and increased BBB permeability. Our BBB transport study showed that compound 1 is a substrate of PYSOCA. Thus, our findings suggest that the hybrid method of HDAC inhibitor and substrate of PYSOCA such as pyrilamine is useful for development of HDAC inhibitors with increased BBB permeability.

16.
J Neurosci ; 38(24): 5584-5595, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29899035

ABSTRACT

Prostaglandin E2 (PGE2) in the hypothalamus is a principal mediator of the febrile response. However, the role of organic anion transporting polypeptide 2A1 (OATP2A1/SLCO2A1), a prostaglandin transporter, in facilitating this response is unknown. Here, we investigated the effect of Slco2a1 deficiency on the body core temperature (Tc) and on the PGE2 concentration in hypothalamus interstitial fluid (Cisf) and CSF (Ccsf) of lipopolysaccharide (LPS; 100 µg/kg, i.p.)-treated mice of both sexes. Slco2a1-/- mice did not develop a febrile response. Ccsf was increased in Slco2a1+/+ and Slco2a1-/- mice, and Ccsf of Slco2a1-/- mice was well maintained at 5 h after LPS injection (1160 pg/ml) compared with Slco2a1+/+ mice (316 pg/ml). A microdialysis study revealed that Cisf peaked at 2 h after LPS injection in Slco2a1+/+ mice (841 pg/ml), whereas the increase in Cisf was negligible in Slco2a1-/- mice. The PGE2 plasma concentration in Slco2a1-/- mice (201 pg/ml) was significantly higher than that in Slco2a1+/+ mice (54 pg/ml) at 1 h after LPS injection, whereas the two groups showed similar PGE2 concentrations in the hypothalamus. Strong Oatp2a1 immunoreactivity was observed in F4/80-positive microglia and perivascular cells and in brain capillary endothelial cells. The changes in Tc and Cisf seen in LPS-injected Slco2a1+/+ mice were partially attenuated in monocyte-/macrophage-specific Slco2a1-/- (Slco2a1Fl/Fl/LysMCre/+) mice. Thus, OATP2A1 facilitates the LPS-induced febrile response by maintaining a high level of Cisf, possibly by regulating PGE2 secretion from F4/80-positive glial cells and/or facilitating PGE2 transport across the blood-brain barrier. These findings suggest that OATP2A1 is a useful therapeutic target for neuroinflammation.SIGNIFICANCE STATEMENT Fever is a physiological response caused by pyrogen-induced release of prostaglandin E2 (PGE2) in the hypothalamus, which plays a central role in regulating the set-point of body temperature. However, it is unclear whether the prostaglandin transporter OATP2A1/SLCO2A1 is involved in this response. We show here that LPS-induced fever is associated with increased PGE2 concentration in hypothalamus interstitial fluid (Cisf), but not in CSF (Ccsf), by means of a microdialysis study in global Slco2a1-knock-out mice and monocyte-/macrophage-specific Slco2a1-knock-out mice. The results suggest that OATP2A1 serves as a regulator of Cisf in F4/80-positive glial cells. OATP2A1 was detected immunohistochemically in brain capillary endothelial cells and, therefore, may also play a role in PGE2 transport across the blood-brain barrier.


Subject(s)
Body Temperature Regulation/physiology , Brain/metabolism , Dinoprostone/metabolism , Fever/metabolism , Organic Anion Transporters/metabolism , Animals , Brain/physiopathology , Fever/chemically induced , Fever/physiopathology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout
17.
J Pharm Sci ; 106(9): 2576-2582, 2017 09.
Article in English | MEDLINE | ID: mdl-28454746

ABSTRACT

Varenicline is a selective partial α4ß2 nicotinic acetylcholine receptor agonist, which is used to help achieve smoking cessation. Here, we investigated varenicline transport at the blood-brain barrier by means of in vivo microdialysis, in situ brain perfusion, and brain efflux index measurements in rats, and in vitro uptake studies in human brain capillary endothelial cells. Microdialysis demonstrated that varenicline is actively transported from blood to brain in rats. Blood-to-brain uptake transport of varenicline, as measured by the in situ brain perfusion technique, was strongly inhibited by diphenhydramine, a potent inhibitor of proton-coupled organic cation (H+/OC) antiporter. However, brain efflux index study showed that brain-to-blood efflux transport of varenicline was not inhibited by diphenhydramine. In human brain capillary endothelial cells, varenicline was taken up time- and concentration-dependently. The uptake was dependent on an oppositely directed proton gradient, but was independent of extracellular sodium and membrane potential. The uptake was inhibited by a metabolic inhibitor, and by substrates of H+/OC antiporter, but not by substrates or inhibitors of OCTs, OCTNs, PMAT, and MATE1, which are known organic cation transporters. The present results suggest that the H+/OC antiporter contributes predominantly to varenicline uptake at the blood-brain barrier.


Subject(s)
Antiporters/metabolism , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Nicotinic Agonists/metabolism , Receptors, Nicotinic/metabolism , Varenicline/metabolism , Animals , Antiporters/chemistry , Biological Transport , Brain/drug effects , Brain/metabolism , Diphenhydramine/chemistry , Diphenhydramine/metabolism , Endothelium, Vascular/metabolism , Humans , Kinetics , Male , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/chemistry , Organic Cation Transport Proteins/metabolism , Protons , Rats , Thermodynamics , Varenicline/administration & dosage , Varenicline/chemistry
18.
J Neurochem ; 139(3): 408-418, 2016 11.
Article in English | MEDLINE | ID: mdl-27501284

ABSTRACT

The elimination of histamine, an excitatory neurotransmitter, from the brain/CSF across the blood-brain barrier and blood-CSF barrier (BCSFB) was investigated using Wistar rats, which were anesthetized with pentobarbital sodium. An in vivo intracerebral microinjection study suggested that there was only partial efflux of [3 H]histamine from the rat brain across the blood-brain barrier. The [3 H]histamine elimination clearance from the rat CSF was 3.8-fold greater than that of a CSF bulk flow marker, and the elimination of [3 H]histamine was significantly inhibited by the co-administration of unlabeled histamine, suggesting the involvement of a carrier-mediated process in histamine elimination from the CSF. The uptake study of [3 H]histamine by the isolated rat choroid plexus revealed histamine elimination from the CSF across the BCSFB. The [3 H]histamine uptake by TR-CSFB3 cells, a model cell line for the BCSFB, exhibited temperature-dependent and saturable kinetics, suggesting the involvement of carrier-mediated transport of histamine at the BCSFB. In the inhibition study, [3 H]histamine uptake by TR-CSFB3 cells was significantly inhibited by substrates and/or inhibitors of plasma membrane monoamine transporter (PMAT/SLC29A4), but not affected by substrates of organic cation transporters. These results suggest the elimination of histamine from the CSF via plasma membrane monoamine transporter at the BCSFB.


Subject(s)
Blood-Nerve Barrier/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Histamine/cerebrospinal fluid , Animals , Blood-Brain Barrier/metabolism , CHO Cells , Cell Membrane/metabolism , Choroid Plexus/metabolism , Cricetinae , Cricetulus , Equilibrative Nucleoside Transport Proteins , Histamine/administration & dosage , Histamine/pharmacology , In Vitro Techniques , Injections, Intraventricular , Lopinavir/pharmacology , Male , Microinjections , Protease Inhibitors/pharmacology , Rats , Rats, Wistar
19.
J Pharm Sci ; 105(2): 935-940, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26554532

ABSTRACT

Brain microdialysis is a powerful tool to estimate brain-to-plasma unbound concentration ratio at the steady state (Kp,uu) of compounds by direct measurement of the unbound concentration in brain interstitial fluid. Here, we evaluated a method to estimate Kp,uu values of multiple organic-cationic drugs simultaneously, by means of brain microdialysis combined with cocktail dosing. Five cationic drugs (diphenhydramine, memantine, oxycodone, pyrilamine, and tramadol), substrates of the proton-coupled organic cation antiport system, were selected as model drugs, and compared under single-dosing and cocktail-dosing conditions. We selected doses of the drugs at which no significant drug-drug interaction occurs at the proton-coupled organic cation antiport system in the blood-brain barrier (BBB). This was confirmed by uptake studies in hCMEC/D3 cells, an in vitro BBB model. The Kp,uu values after cocktail administration were in the range of 1.8-5.2, and were in good agreement with those after single administration. These results suggest that the microdialysis method with cocktail dosing is suitable to estimate Kp,uu values of several cationic drugs simultaneously, if there is no drug-drug interaction during BBB transport. The method could be useful for evaluating drug candidates with high Kp,uu values at an early stage in the development of central nervous system-acting drugs.


Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Microdialysis/methods , Pharmaceutical Preparations/metabolism , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Cell Line, Transformed , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Male , Pharmaceutical Preparations/administration & dosage , Rats , Rats, Wistar
20.
Yakugaku Zasshi ; 135(5): 697-702, 2015.
Article in Japanese | MEDLINE | ID: mdl-25948304

ABSTRACT

The transport of opioid analgesics across the blood-brain barrier (BBB) is an important determinant of their therapeutic effects. The human brain is protected by the BBB, which consists of brain capillary endothelial cells linked with tight junctions. It is well established that the polarized expression of numerous transporters and receptors at the brain capillary endothelial cells controls the blood-brain exchange of nutrients, waste products deriving from neurotransmitter substances, and drugs. Morphine is a substrate of P-glycoprotein and the P-glycoprotein-mediated efflux transport at the BBB maintains a lower unbound concentration of morphine in the brain compared with plasma. On the other hand, oxycodone has 3 times higher unbound concentration in the brain than plasma, suggesting an active transport mechanism of oxycodone across the BBB into the brain. In vitro transport study using BBB model cells showed that oxycodone is efficiently transported by a proton-coupled organic cation antiporter. Human BBB model cells also retain the proton-coupled organic cation antiporter. Although adjuvant analgesics include many cationic drugs that interact with oxycodone transport across the BBB at relatively high concentrations, these drugs would enhance the antinociceptive effects of oxycodone with little effect on oxycodone pharmacokinetics, including brain distribution at therapeutically or pharmacologically relevant concentrations. These findings support the idea that proton-coupled organic cation antiporter-mediated transport of oxycodone at the BBB plays a role in determining the therapeutic efficacy of this opioid analgesic drug.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiology , Brain/metabolism , Oxycodone/pharmacokinetics , Analgesics, Opioid/metabolism , Animals , Antiporters/metabolism , Antiporters/physiology , Biological Transport , Humans , Oxycodone/metabolism , Rats , Therapeutic Equivalency
SELECTION OF CITATIONS
SEARCH DETAIL