Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 38(1): 168-76, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19833843

ABSTRACT

This study investigated the role of a multispecific organic anion transporter, Oatp1a4/Slco1a4, in drug transport across the blood-brain barrier. In vitro transport studies using human embryonic kidney 293 cells expressing mouse Oatp1a4 identified the following compounds as Oatp1a4 substrates: pitavastatin (K(m) = 8.3 microM), rosuvastatin (K(m) = 12 microM), pravastatin, taurocholate (K(m) = 40 microM), digoxin, ochratoxin A, and [d-penicillamine(2,5)]-enkephalin. Double immunohistochemical staining of Oatp1a4 with P-glycoprotein (P-gp) or glial fibrillary acidic protein demonstrated that Oatp1a4 signals colocalized with P-gp signals partly but not with glial fibrillary acidic protein, suggesting that Oatp1a4 is expressed in both the luminal and the abluminal membranes of mouse brain capillary endothelial cells. The brain-to-blood transport of pitavastatin, rosuvastatin, pravastatin, and taurocholate after microinjection into the cerebral cortex was significantly decreased in Oatp1a4(-/-) mice compared with that in wild-type mice. The blood-to-brain transport of pitavastatin, rosuvastatin, taurocholate, and ochratoxin A, determined by in situ brain perfusion, was significantly lower in Oatp1a4(-/-) mice than in wild-type mice, whereas transport of pravastatin and [D-penicillamine(2,5)]-enkephalin was unchanged. The blood-to-brain transport of digoxin was significantly lower in Oatp1a4(-/-) mice than in wild-type mice only when P-gp was inhibited by N-(4-[2-(1,2,3,4-tetrahydro-6,7-dimethoxy-2-isoquinolinyl)ethyl]-phenyl)-9,10-dihydro-5-methoxy-9-oxo-4-acridine carboxamide (GF120918). Taken together, these results show that Oatp1a4 can mediate the brain-to-blood and blood-to-brain transport of its substrate drugs across the blood-brain barrier. The brain-to-plasma ratio of taurocholate, pitavastatin, and rosuvastatin was close to the capillary volume in wild-type mice, and it was not affected by Oatp1a4 dysfunction. Whether Oatp1a4 can deliver drugs from the blood to the brain remains controversial.


Subject(s)
Blood-Brain Barrier/metabolism , Organic Anion Transporters/metabolism , Organic Cation Transport Proteins/metabolism , Pharmaceutical Preparations/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Acridines/pharmacology , Animals , Blood-Brain Barrier/drug effects , Brain/blood supply , Brain/drug effects , Brain/metabolism , Capillaries/metabolism , Cell Line , Cell Membrane/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Choroid Plexus/blood supply , Choroid Plexus/metabolism , Digoxin/administration & dosage , Digoxin/metabolism , Digoxin/pharmacokinetics , Enkephalin, D-Penicillamine (2,5)-/administration & dosage , Enkephalin, D-Penicillamine (2,5)-/metabolism , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Fluorobenzenes/administration & dosage , Fluorobenzenes/blood , Fluorobenzenes/metabolism , Fluorobenzenes/pharmacokinetics , Gene Expression/genetics , Humans , Ion Pumps/genetics , Kinetics , Liver/blood supply , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ochratoxins/administration & dosage , Ochratoxins/metabolism , Ochratoxins/pharmacokinetics , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Pravastatin/administration & dosage , Pravastatin/metabolism , Pravastatin/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/blood , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics , Quinolines/administration & dosage , Quinolines/blood , Quinolines/metabolism , Quinolines/pharmacokinetics
2.
Neuropharmacology ; 55(5): 755-62, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18602932

ABSTRACT

The highly selective delta opioid agonist, SNC80, elicits dopamine-related behaviors including locomotor stimulation and conditioned place-preference. In contrast, it has been reported that SNC80 fails to promote dopamine efflux from the striatum of freely moving rats. However, SNC80 does enhance behavioral responses to the stimulants, amphetamine and cocaine, suggesting an interaction between delta opioids and psychostimulants. Since the increase in locomotor activity elicited by amphetamine and related stimulants acting at the dopamine transporter is associated with increases in extracellular concentrations of dopamine within the striatum, we hypothesized that SNC80 enhances this activity by potentiating the overflow of dopamine through the transporter. To test this hypothesis, striatal preparations from Sprague Dawley rats were assayed for dopamine efflux in response to amphetamine challenge. SNC80 was given either in vivo or in vitro directly to rat striatal tissue, prior to in vitro amphetamine challenge. Both in vivo and in vitro administration of SNC80 enhanced amphetamine-mediated dopamine efflux in a concentration- and time-dependent manner. However, SNC80 in either treatment paradigm produced no stimulation of dopamine efflux in the absence of amphetamine. The effect of SNC80 on amphetamine-mediated dopamine overflow, but not the effect of amphetamine alone, was blocked by the delta selective antagonist, naltrindole and was also observed with other delta agonists. The results of this study demonstrate that even though SNC80 does not stimulate dopamine efflux alone, it is able to augment amphetamine-mediated dopamine efflux through a delta opioid receptor mediated action locally in the striatum.


Subject(s)
Amphetamine/pharmacology , Benzamides/pharmacology , Corpus Striatum/drug effects , Dopamine Uptake Inhibitors/pharmacology , Dopamine/metabolism , Piperazines/pharmacology , Receptors, Opioid, delta/agonists , Analgesics, Opioid/pharmacokinetics , Analysis of Variance , Animals , Area Under Curve , Dose-Response Relationship, Drug , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Male , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/physiology , Subcellular Fractions/drug effects , Time Factors , Tritium/pharmacokinetics
3.
Brain Res Bull ; 64(6): 471-9, 2005 Jan 30.
Article in English | MEDLINE | ID: mdl-15639542

ABSTRACT

The occurrence of systematic diurnal variations in pain thresholds has been demonstrated in human. Salivary melatonin levels change following acute pain when other factors that could explain the change have been removed or controlled. Melatonin-induced analgesia is blocked by naloxone or pinealectomy. By using selective radioligands [3H]-DAMGO, [3H]-DPDPE, [3-U69593, and 3H]-nociceptin, we have shown that the bovine pinealocytes contain delta and mu, but not kappa or ORL1 opioid receptor subtypes. In the present study, by using melatonin receptor agonists (6-chloromelatonin or 2-iodo-N-butanoyl-5-methoxytryptamine) or melatonin receptor antagonist (2-phenylmelatonin), we have shown that these agents do not compete with opioid receptor subtypes. However, we observed a time-dependent release of beta-endorphin an endogenous opioid peptide, by melatonin from mouse pituitary cells in culture. Hence, it is suggested that melatonin exerts its analgesic actions not by binding to opioid receptor subtypes but by binding to its own receptors and increasing the release of beta-endorphin.


Subject(s)
Analgesics/pharmacology , Melatonin/analogs & derivatives , Melatonin/pharmacology , Pineal Gland/cytology , Receptors, Opioid/metabolism , beta-Endorphin/metabolism , Analgesics, Opioid/pharmacokinetics , Animals , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/cytology , Brain/drug effects , Brain/metabolism , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Enzyme-Linked Immunosorbent Assay/methods , Melatonin/agonists , Melatonin/antagonists & inhibitors , Melatonin/chemistry , Mice , Naloxone/chemistry , Naloxone/pharmacology , Opioid Peptides/pharmacokinetics , Pineal Gland/metabolism , Radioligand Assay , Rats , Receptors, Opioid/agonists , Receptors, Opioid/classification , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Time Factors , Tritium/pharmacokinetics , Nociceptin
4.
J Pharmacol Exp Ther ; 311(3): 1203-10, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15302892

ABSTRACT

[D-Pen2,D-Pen5]-Enkephalin (DPDPE) is excreted extensively into the bile. Although DPDPE is transported by P-glycoprotein (P-gp), multidrug resistance-associated protein 2 (Mrp2) has been identified as an important mechanism for DPDPE transport across the canalicular membrane of the hepatocyte. The present studies determined the relative impact of Mrp2 and P-gp on the hepatobiliary disposition of [3H]DPDPE in isolated perfused rat livers (IPLs). Perfusate clearance of [3H]DPDPE was not different between livers from control and Mrp2-deficient (TR-) rats. Biliary excretion of [3H]DPDPE in IPLs from Wistar control rats was rapid and extensive. However, when [3H]DPDPE was administered to livers from TR- rats, the rate and extent of excretion decreased significantly. Surprisingly, in the presence of the P-gp inhibitor GF120918 [N-(4-[2-(1,2,3,4-tetrahydro-6,7-dimethoxy-2-isoquinolinyl)ethyl]-phenyl)-9,10-dihydro-5-methoxy-9-oxo-4-acridine carboxamide], biliary excretion of [3H]DPDPE was not inhibited in control livers. In contrast, administration of GF120918 to TR- livers further reduced the maximal excretion rate and decreased net biliary excretion of [3H]DPDPE by 87%. GF120918 administration caused an unexpected increase in perfusate clearance in both control and TR- rat livers. At distribution equilibrium, [3H]DPDPE liver/perfusate partitioning was higher in GF120918-treated livers. Results of pharmacokinetic modeling were consistent with the hypothesis that GF120918 inhibited a [3H]DPDPE basolateral excretion mechanism. Mrp2 is the primary mechanism for [3H]DPDPE biliary excretion, and P-gp facilitates excretion of [3H]DPDPE only in the absence of functional Mrp2. [3H]DPDPE is a substrate for a basolateral efflux mechanism that is sensitive to inhibition by GF120918. These data emphasize the importance of using appropriate model systems and comprehensive pharmacokinetic modeling in elucidating the complex interplay between multiple transport systems.


Subject(s)
Bile/metabolism , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Liver/metabolism , Acridines/pharmacology , Animals , Area Under Curve , Carrier Proteins/metabolism , Hepatocytes/metabolism , In Vitro Techniques , Male , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Models, Biological , Nonlinear Dynamics , Rats , Rats, Wistar , Ribosomal Proteins/deficiency , Ribosomal Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Tetrahydroisoquinolines/pharmacology
5.
Pharm Res ; 21(7): 1294-302, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15290872

ABSTRACT

PURPOSE: The isolation of hepatocytes from intact liver involves collagenase digestion of the tissue, resulting in loss of cell polarization and functional vectorial excretion. These studies examined repolarization, localization of P-glycoprotein (P-gp) to the canalicular domain of the hepatocyte, and re-establishment of vectorial transport in sandwich-cultured (SC) rat and human primary hepatocytes. METHODS: Protein localization and expression were determined in SC hepatocytes by confocal microscopy and Western blotting, respectively. Transporter function was evaluated by measuring [D-penicillamine2,5]enkephalin (3H-DPDPE) and 5 (and 6)-carboxy-2',7'-dichlorofluorescein (CDF) biliary excretion in SC hepatocytes. RESULTS: P-gp and the canalicular marker protein dipeptidyl peptidase IV (DPPIV) co-localized by Day 3 and Day 6 in SC rat hepatocytes and SC human hepatocytes, respectively, consistent with canalicular network formation visualized by light microscopy. Co-localization of multidrug resistance associated protein 2 (MRP2) and P-gp in SC human hepatocytes was observed on Day 6 in culture. Expression levels of P-gp increased slightly in both species over days in culture; similar expression was observed for MRP2 in SC human hepatocytes. Oatp1a1 expression in SC rat hepatocytes was maintained over days in culture, whereas Oatp1a4 expression decreased. OATP1B1 expression decreased slightly on Day 3 in SC human hepatocytes. OATP1B3 expression was constant in SC human hepatocytes. In vitro biliary excretion of the opioid peptide 3H-DPDPE correlated with the proper localization of canalicular proteins in both species. Excretion of CDF in SC human hepatocytes confirmed network formation and MRP2 function. CONCLUSIONS: These studies indicate that SC hepatocytes repolarize and traffic functional canalicular transport proteins to the appropriate cellular domain.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Bile/metabolism , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Hepatocytes/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Bile/drug effects , Biological Transport , Blotting, Western , Cells, Cultured , Dipeptidyl Peptidase 4/metabolism , Fluorescent Antibody Technique , Hepatocytes/drug effects , Humans , Male , Membrane Transport Proteins/metabolism , Microscopy, Confocal , Models, Biological , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transporters/metabolism , Rats , Rats, Wistar , Species Specificity
6.
J Pharm Pharmacol ; 55(7): 1013-20, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12906759

ABSTRACT

The objective of this study was to examine the transport activity of the human organic anion transporter OATP-C (SLC21A6) for oligopeptides that are eliminated rapidly from the systemic circulation. We focused on an opioid peptide analogue, [D-Ala(2), D-Leu(5)]-enkephalin (DADLE), a linear pentapeptide modified to be stable. [(3)H]DADLE was taken up by rat isolated hepatocytes in a saturable manner and highly accumulated in the liver after intravenous administration to rats. The uptake of [(3)H]DADLE by the isolated hepatocytes was inhibited by several organic anions and pentapeptides, but not by tetra- or tripeptides. When OATP-C was expressed in Xenopus laevis oocytes, a significant increase in uptake of [(3)H]DADLE was observed. Moreover, the inhibitory effects of various compounds, including some peptides, on [(3)H]estrone-3-sulfate uptake by OATP-C were similar to those observed in [(3)H]DADLE uptake by rat isolated hepatocytes. In conclusion, it was demonstrated that OATP-C contributes to the rapid hepatic excretion of peptides and peptide-mimetic drugs.


Subject(s)
Enkephalin, Leucine-2-Alanine/pharmacokinetics , Estrone/analogs & derivatives , Hepatocytes/metabolism , Liver-Specific Organic Anion Transporter 1/metabolism , Animals , Anions , Bile , Biological Transport , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Estrone/pharmacokinetics , In Vitro Techniques , Injections, Intravenous , Oocytes/metabolism , Peptides/metabolism , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Xenopus laevis
7.
Arzneimittelforschung ; 53(1): 21-5, 2003.
Article in English | MEDLINE | ID: mdl-12608010

ABSTRACT

Imipramine (CAS 113-52-0) is being utilized widely for the treatment of major depression. In recent years, there has been evidence of the involvement of the endogenous opioid system in major depression and its treatment. There is some evidence indicating that opioid receptors could be involved in the antidepressant mechanism of action. Regarding this topic, mood-related behavior of endogenous enkephalins seems to be mediated by delta-opioid receptors. In this work, the effects of subacute (5 day) and chronic (15 day) treatments of imipramine on the density and the affinity of the delta-receptors in the striatum and in the parietal and frontal cortices of the rat brain are described. Studied parameters (Bmax and Kd) were calculated by a saturation binding assay with the delta-opioid agonists [3H]-DPDPE (tyrosyl-2,6-3H(N)-(2-D-penicillamine-5-D-penicillamine)-enkephalin) as specific ligand and DSLET ([D-serine2]-D-leucine-enkephalin-threonine) as non-radioactive competing ligand. It was found that 15 days treatment significantly decreased the delta-opioid receptor density,without changing the affinity, in the frontal cortex of the rat brain. That decrease was confirmed by delta-opioid receptor immunostaining. These results suggest that delta-opioid receptors could play a role in the chronic action mechanism of imipramine.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Cerebral Cortex/metabolism , Imipramine/pharmacology , Neostriatum/metabolism , Receptors, Opioid, delta/drug effects , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacokinetics , Animals , Binding, Competitive/drug effects , Cerebral Cortex/drug effects , Enkephalin, D-Penicillamine (2,5)-/metabolism , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Immunohistochemistry , Male , Neostriatum/drug effects , Rats , Rats, Sprague-Dawley
8.
Toxicol Sci ; 71(2): 263-75, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12563112

ABSTRACT

To define the effects of chronic spinal exposure to a highly selective partial differential opioid agonist c[DPen(2),DPen(5)]enkephalin (DPDPE), adult beagles were prepared with chronic lumbar intrathecal catheters. Groups of dogs received intrathecal infusions (100 micro l/h) of saline (vehicle), DPDPE 3 mg/ml or 6 mg/ml for 28 days. Over the 28-day period, saline or 3 mg/ml showed minimal changes in neurological function, whereas in the 6 mg/ml animals, prominent hind limb dysfunction evolved over the 28-day interval. Histopathology in control animals displayed a modest pericatheter reaction considered normal for this model. Dogs receiving DPDPE (three of four at 6 mg/ml and one of four at 3 mg/ml) but not saline (zero of four) developed large inflammatory masses (granulomas) in the intrathecal space located proximal to the catheter tip. In these masses, severe chronic inflammatory changes in combination with necrosis and fibrosis was detected. Occasional focal destruction of neuropil was detected also in the adjacent spinal cord parenchyma. These masses contained extensive accumulation of mouse antihuman macrophages (MAC)-positive inflammatory cells expressing tumor necrosis factor-alpha (TNF-alpha), revealing infiltration of macrophages, granulocytes, and monocytes. In separate animals, prepared with dual intrathecal catheters, lumbar CSF was sampled at specified time points following intrathecal bolus (3 mg/ml) and 24 h DPDPE infusion (3 mg/ml and 6 mg/ml). Steady-state cerebrospinal fluid (CSF) DPDPE levels were 18.6 +/- 1.0 and 22.6 +/- 4.0 micro g/ml for 3 mg/ml and 6 mg/ml infusions respectively. These results indicate that this partial differential opioid agonist DPDPE produces a concentration and time-dependent formation of an intrathecal inflammatory mass.


Subject(s)
Analgesics, Opioid/toxicity , Behavior, Animal/drug effects , Enkephalin, D-Penicillamine (2,5)-/toxicity , Granuloma/chemically induced , Spinal Cord Diseases/chemically induced , Analgesics, Opioid/administration & dosage , Analgesics, Opioid/pharmacokinetics , Animals , Area Under Curve , Catheters, Indwelling , Dogs , Dose-Response Relationship, Drug , Enkephalin, D-Penicillamine (2,5)-/administration & dosage , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Female , Granuloma/pathology , Injections, Spinal , Male , Motor Activity/drug effects , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord Diseases/pathology
9.
BMC Neurosci ; 3: 19, 2002 Nov 18.
Article in English | MEDLINE | ID: mdl-12437765

ABSTRACT

BACKGROUND: The aim of the present study was to characterize the activation profiles of 15 opioid ligands in transfected human embryonic kidney cells expressing only delta opioid receptors. Activation profiles of most of these ligands at delta opioid receptors had not been previously characterized in vitro. Receptor activation was assessed by measuring the inhibition of forskolin-stimulated cAMP production. RESULTS: Naltrexone and nalorphine were classified as antagonists at delta opioid receptor. The other ligands studied were agonists at delta opioid receptors and demonstrated IC50 values of 0.1 nM to 2 microM, maximal inhibition of 39-77% and receptor binding affinities of 0.5 to 243 nM. The rank order of efficacy of the ligands tested was metazocine = xorphanol > or = fentanyl = SKF 10047 = etorphine = hydromorphone = butorphanol = lofentanil > WIN 44,441 = Nalbuphine = cyclazocine > or = met-enkephalin >> morphine = dezocine. For the first time these data describe and compare the function and relative efficacy of several ligands at delta opioid receptors. CONCLUSIONS: The data produced from this study can lead to elucidation of the complete activation profiles of several opioid ligands, leading to clarification of the mechanisms involved in physiological effects of these ligands at delta opioid receptors. Furthermore, these data can be used as a basis for novel use of existing opioid ligands based on their pharmacology at delta opioid receptors.


Subject(s)
Kidney/metabolism , Receptors, Opioid, delta/metabolism , Animals , Binding, Competitive , Cell Line , Colforsin/pharmacology , Cyclic AMP/metabolism , Dose-Response Relationship, Drug , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Humans , Kidney/cytology , Kidney/drug effects , Ligands , Mice , Narcotic Antagonists/pharmacology , Narcotics/pharmacology , Receptors, Opioid, delta/drug effects , Receptors, Opioid, delta/genetics , Transfection
10.
Article in English | MEDLINE | ID: mdl-12016017

ABSTRACT

An atmospheric pressure ionization liquid chromatographic-mass spectrometric assay was developed and validated for the determination of D-penicillamine(2,5) enkephalin (DPDPE) in cerebrospinal fluid (CSF) from dog. DPDPE and internal standard (D-Ala(2),D-Leu(5) enkephalin=DADLE) were isolated from CSF by reversed-phase C(18) solid-phase extraction with ZipTip micro-cartridges. Aliquots of extracted eluate were injected onto an Agilent Zorbax SB C(18) column (30 x 2.2 mm; 3.5 microm) at a flow-rate of 0.4 ml/min. The isocratic mobile phase of methanol-10 mM ammonium formate (pH 3) (75:25, v/v) was then diverted to waste for 45 s after injection, after which time flow was directed to the single quadrupole mass spectrometer. DPDPE was detected by positive mode selected ion monitoring. Standard curves were linear (r(2)> or =0.991) over the concentration range 1-1000 ng/ml. The efficiency of extraction recovery was greater than 97%, and the intra-assay and inter-assay precisions were within 9% relative standard deviation. DPDPE and the internal standard were stable in the injection solvent at 4 degrees C for at least 48 h. The assay was applied to the pharmacokinetic study of intrathecal DPDPE administration in the dog animal model.


Subject(s)
Analgesics, Opioid/cerebrospinal fluid , Chromatography, High Pressure Liquid/methods , Enkephalin, D-Penicillamine (2,5)-/cerebrospinal fluid , Mass Spectrometry/methods , Analgesics, Opioid/pharmacokinetics , Animals , Dogs , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
11.
Int J Pharm ; 233(1-2): 85-98, 2002 Feb 21.
Article in English | MEDLINE | ID: mdl-11897413

ABSTRACT

In keeping with the advance of biotechnology, cell culture becomes an important tool for investigating the transport and the metabolism phenomena. A cell line of human origin, the BeWo choriocarcinoma cell line, was used for the study of the transport and metabolism of opioid peptides across the in vitro model of the placental barrier. Opioid peptides, both naturally occurring and their synthetic analogs, are of interest to be developed as potent analgesics and were included in this study. The apparent permeability coefficients (Pe)s of the peptides containing 4-11 amino acid or analog residues were in the range of 0.23-14.6 x 10(-5) cm/s. The (Pe)s of these peptides were comparable to those of sucrose or dextrans, hydrophilic markers. The (Pe)s of low molecular weight (MW) peptides was not dependent on their MW or molecular size, whereas an inversely linear correlation between (Pe)s and molecular size was observed with the larger peptides. Molecular sieving of the BeWo monolayer restricted the transport of the peptides with MW> or =1033 Da or molecular size > or =6.6 A. Membrane partitioning ability and charge of the peptides were also investigated and found to be the minor factors regulating the extent of peptide permeation. Contrasting to the transport of Tyr-[D-pen-Gly-Phe-D-Pen] (DPDPE) peptide analog across the blood-brain barrier, the transport of DPDPE across the BeWo monolayers were not indicated to be via carrier-mediated transport. The major transport pathway of the opioid peptides across the BeWo monolayers was found to be via paracellular route. In metabolism studies, aminopeptidase was found to be a major enzyme type responsible for the degradation of naturally occurring peptides but not for the synthetic analogs. The finding obtained from the present study reveals the applicability of the BeWo cell line as an in vitro model for investigating placental transport and metabolism of opioid peptides.


Subject(s)
Opioid Peptides/metabolism , Placenta/metabolism , Tumor Cells, Cultured/metabolism , Analgesics, Opioid/pharmacokinetics , Animals , Biological Transport/physiology , Choriocarcinoma/metabolism , Drug Stability , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Female , Humans , Molecular Weight , Rats , Uterine Neoplasms/metabolism
12.
Mol Pharmacol ; 59(4): 774-83, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259622

ABSTRACT

mu- and delta-Opioid agonists interact in a synergistic manner to produce analgesia in several animal models. Additionally, receptor binding studies using membranes derived from brain tissue indicate that interactions between mu- and delta-opioid receptors might be responsible for the observation of multiple opioid receptor subtypes. To examine potential interactions between mu- and delta-opioid receptors, we examined receptor binding and functional characteristics of mu-, delta-, or both mu- and delta-opioid receptors stably transfected in rat pituitary GH(3) cells (GH(3)MOR, GH(3)DOR, and GH(3)MORDOR, respectively). Saturation and competition binding experiments revealed that coexpression of mu- and delta-opioid receptors resulted in the appearance of multiple affinity states for mu- but not delta-opioid receptors. Additionally, coadministration of selective mu- and delta-opioid agonists in GH(3)MORDOR cells resulted in a synergistic competition with [(3)H][D-Pen(2,5)]enkephalin (DPDPE) for delta-opioid receptors. Finally, when equally effective concentrations of [D-Ala(2),N-MePhe(4),Gly-ol(5)]enkephalin (DAMGO) and two different delta-opioid agonists (DPDPE or 2-methyl-4a alpha-(3-hydroxyphenyl)-1,2,3,4,4a,5,12,12a alpha-octahydroquinolino-[2,3,3-g]-isoquinoline; TAN67) were coadministered in GH(3)MORDOR cells, a synergistic inhibition of adenylyl cyclase activity was observed. These results strongly suggest that cotransfection of mu- and delta-opioid receptors alters the binding and functional characteristics of the receptors. Therefore, we propose that the simultaneous exposure of GH(3)MORDOR cells to selective mu- and delta-opioid agonists produces an interaction between receptors resulting in enhanced receptor binding. This effect is translated into an augmented ability of these agonists to inhibit adenylyl cyclase activity. Similar interactions occurring in neurons that express both mu- and delta-opioid receptors could explain observations of multiple opioid receptor subtypes in receptor binding studies and the synergistic interaction of mu- and delta-opioids in analgesic assays.


Subject(s)
Pituitary Gland/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Adenylyl Cyclase Inhibitors , Analgesics, Opioid/metabolism , Analgesics, Opioid/pharmacokinetics , Animals , Binding, Competitive/drug effects , Cell Line , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Enkephalin, D-Penicillamine (2,5)-/metabolism , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Gene Expression , Pituitary Gland/cytology , Quinolines/pharmacology , Rats , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics , Transfection
13.
Neurosci Lett ; 301(3): 155-8, 2001 Apr 06.
Article in English | MEDLINE | ID: mdl-11257421

ABSTRACT

P-glycoprotein (P-gp) and organic anion transporting polypeptides (Oatp) are expressed at the blood-brain barrier (BBB). There is little functional evidence for Oatp-mediated transport at the BBB. The peptidic delta opioid-receptor agonist [D-penicillamine(2,5)]-enkephalin (DPDPE) is a substrate of mdr1a P-gp and Oatp2. The present study evaluated the influence of these transporters on brain uptake of DPDPE by in situ perfusion in mice. Brain uptake was increased approximately 12-fold in mice lacking P-gp in the BBB, but the P-gp inhibitor dexverapamil did not increase uptake in P-gp-competent mice. In P-gp-deficient mice, DPDPE uptake was saturable (K(m) approximately 24 mM), and was inhibited by dexverapamil and the Oatp2 substrates digoxin, estradiol-17beta-glucuronide and fexofenadine. These results confirm P-gp-mediated efflux of DPDPE, and suggest functional uptake transport of DPDPE by Oatp, at the murine BBB.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , ATP-Binding Cassette Transporters/genetics , Analgesics, Opioid/pharmacokinetics , Blood-Brain Barrier/physiology , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Receptors, Opioid, delta/metabolism , Terfenadine/analogs & derivatives , Animals , Anion Transport Proteins , Blood-Brain Barrier/drug effects , Calcium Channel Blockers/pharmacology , Carrier Proteins/metabolism , Digoxin/pharmacology , Enzyme Inhibitors/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Histamine H1 Antagonists/pharmacology , Male , Mice , Mice, Knockout , Perfusion , Receptors, Opioid, delta/agonists , Terfenadine/pharmacology , Verapamil/pharmacology
14.
Nat Neurosci ; 4(3): 268-74, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11224543

ABSTRACT

Many peptides and transmitters found within the brain also have peripheral sites of action. We now demonstrate that the brain releases functionally active neurotransmitters/neuromodulators directly from the brain into the blood through a saturable P-glycoprotein (Pgp) transport system. Downregulating Pgp1 expression with antisense reduced the brain-to-blood transport of morphine, beta-endorphin and other opioids. Lowering Pgp expression significantly enhanced systemic morphine analgesia and prevented tolerance, but diminished the analgesic activity of centrally administered morphine, implying that supraspinal analgesia resulted from a combination of central and peripheral mechanisms activated by morphine transported from the brain to the blood. Similarly, mice with a disruption of the Mdr1a gene were more sensitive to systemic morphine and less sensitive to morphine given centrally. This ability of the Pgp transport system to pump functionally active compounds from the brain to periphery defines a potentially important mechanism for the central nervous system to modulate peripheral systems.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Blood-Brain Barrier/physiology , Brain/metabolism , Narcotics/blood , Narcotics/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/drug effects , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Biological Transport/drug effects , Biological Transport/physiology , Blood-Brain Barrier/drug effects , Brain/drug effects , Down-Regulation/drug effects , Down-Regulation/physiology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Male , Mice , Mice, Knockout , Morphine/pharmacokinetics , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , beta-Endorphin/pharmacokinetics
15.
J Pharmacol Exp Ther ; 295(3): 972-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11082431

ABSTRACT

Insulin crosses the blood-brain barrier (BBB) via receptor-mediated transcytosis and has been suggested to augment uptake of peripheral substances across the BBB. The delta-opioid receptor-selective peptide D-penicillamine(2,5) (DPDPE), a Met-enkephalin analog, produces analgesia via a central nervous system-derived effect. In vitro (K(cell), microl. min(-1). mg(-1)) and in situ (K(in), microl. min(-1). g(-1)) analyses of DPDPE transport (K(cell) = 0.56 +/- 0. 15; K(in) = 0.28 +/- 0.03) revealed significant (P <.01) increases in DPDPE uptake by the BBB with 10 microM insulin (K(cell) = 1.61 +/- 0.25; K(in) = 0.48 +/- 0.04). In vitro cellular uptake was significantly increased (P <.05) at 1 microM insulin, whereas no significant uptake was observed with CTAP (a somatostatin opioid peptide analog) or sucrose (a paracellular diffusionary marker). No significant change in uptake was seen with DPDPE, CTAP, or sucrose in the presence of holo-transferrin (0-100 microM), indicating that the effect of insulin on DPDPE was not a generalized effect of receptor endocytosis. Insulin did not affect P-glycoprotein efflux, a mechanism that has shown affinity for DPDPE. A similar uptake of DPDPE into the brain (64% increase) was seen with the in situ brain perfusion model. Analgesic assessment revealed a significant decline in DPDPE (i.v.)-induced analgesia with increasing concentrations of insulin (i.v., i.c.v., s.c.) in a dose-dependent manner. Thus, insulin significantly increases DPDPE uptake across the BBB by a specific mechanism. The analgesic effect seen with DPDPE and insulin coadministration was shown to decrease, indicating that insulin reduces the analgesic effect within the central nervous system rather than at the BBB.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Blood-Brain Barrier/drug effects , Insulin/pharmacology , Analgesics, Opioid/pharmacology , Animals , Biological Transport/drug effects , Brain/metabolism , Cattle , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics
16.
Bioorg Med Chem Lett ; 10(20): 2259-61, 2000 Oct 16.
Article in English | MEDLINE | ID: mdl-11055333

ABSTRACT

The study of kappa-opioid receptor function in vivo has been hampered by the limited choice of selective kappa-antagonists. Recently discovered kappa-antagonists have not yet been utilised in vivo. We here report the synthesis and in vitro evaluation of a new amidine derivative of naltrindole. It showed substantially greater kappa-selectivity in binding assays than known analogues with shorter spacer in the amidine side chain. This indicates that in nor-BNI and related selective kappa-antagonists, the second basic centre may not be ideally located.


Subject(s)
Amidines/chemical synthesis , Benzeneacetamides , Naltrexone/analogs & derivatives , Narcotic Antagonists/chemical synthesis , Receptors, Opioid, kappa/antagonists & inhibitors , Amidines/chemistry , Amidines/pharmacology , Animals , Binding, Competitive , Brain/metabolism , Cell Membrane/metabolism , Drug Design , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacokinetics , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Guinea Pigs , Indicators and Reagents , Kinetics , Naltrexone/chemical synthesis , Naltrexone/chemistry , Naltrexone/pharmacology , Narcotic Antagonists/chemistry , Narcotic Antagonists/pharmacology , Pyrrolidines/pharmacokinetics , Structure-Activity Relationship
17.
J Pharmacol Exp Ther ; 294(1): 73-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10871297

ABSTRACT

Organic anion-transporting polypeptides (Oatps) are a rapidly growing gene family of polyspecific membrane transporters. In rat brain, Oatp1 (gene symbol Slc21a1) and Oatp2 (Slc21a5) are localized at the apical and basolateral domains, respectively, of the choroid plexus epithelium. Furthermore, Oatp2 is strongly expressed at the rat blood-brain barrier (BBB). This study localizes the human OATP (now called OATP-A; SLC21A3) at the BBB in humans. Furthermore, with the Xenopus laevis oocyte system the delta-opioid receptor agonists [D-penicillamine(2,5)]enkephalin (DPDPE) and deltorphin II were identified as new transport substrates of OATP-A. This OATP-A-mediated DPDPE and deltorphin II transport exhibited apparent K(m) values of approximately 202 and 330 microM, respectively, and OATP-A-mediated deltorphin II transport was inhibited by the mu-opioid receptor agonist Tyr-D-Ala-Gly-N-methyl-Phe-glycinol, the endogenous peptide Leu-enkephalin, and the opiate antagonists naloxone and naltrindole. DPDPE also was transported by rat Oatp1 (K(m) approximately 48 microM) and Oatp2 (K(m) approximately 19 microM), whereas deltorphin II was only transported by Oatp1 (K(m) approximately 137 microM). These results demonstrate that OATP-A can mediate transport of the analgesic opioid peptides DPDPE and deltorphin II across the human BBB. Furthermore, because rat Oatp1 and Oatp2 exhibit similar but not identical transport activities as OATP-A, the results generally indicate that members of the Oatp/OATP gene family of membrane transporters play an important role in carrier-mediated transport of opioid peptides across the BBB and blood-cerebrospinal fluid barrier of the mammalian brain.


Subject(s)
Blood-Brain Barrier , Carrier Proteins/physiology , Opioid Peptides/pharmacokinetics , Aged , Animals , Anion Transport Proteins , Biological Transport , Carrier Proteins/analysis , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Humans , Male , Molecular Weight , Oligopeptides/pharmacokinetics , Rats
18.
Peptides ; 20(10): 1229-38, 1999.
Article in English | MEDLINE | ID: mdl-10573295

ABSTRACT

The utility of a drug depends on its ability to reach appropriate receptors at the target tissue and remain metabolically stable to produce the desired effect. To improve central nervous system entry of the opioid analgesic [D-Pen2, L-Pen5, Phe6] Enkephalin (DPLPE-Phe), our research group synthesized analogs that had chloro, bromo, fluoro, and iodo halogens on the para positions of the phenylalanine-4 residue. This study reports on investigation of the effect of halogenation on stability, lipophilicity, and in vitro blood-brain barrier permeability of a novel enkephalin analog DPLPE-Phe. The stability of each halogenated DPLPE-Phe analog as well as the amidated and nonamidated parent peptide was tested in plasma and brain. All peptides tested had a half-time disappearance >300 min except for DPLPE-Phe-NH2, which was found to have a half-life of 30 min in plasma. Octanol/saline distribution studies indicated addition of halogens to DPLPE-Phe-OH significantly increased lipophilicity except for p-[F-Phe4]DPLPE-Phe-OH. p-[Cl-Phe4]DPLPE-Phe-OH exhibited the most pronounced increase in lipophilicity. Para-bromo and para-chloro halogen additions significantly enhanced in vitro blood-brain barrier permeability, providing evidence for improved delivery to the central nervous system.


Subject(s)
Blood-Brain Barrier/physiology , Enkephalin, D-Penicillamine (2,5)-/analogs & derivatives , Enkephalins/pharmacokinetics , Halogens/metabolism , Peptides/pharmacokinetics , Animals , Cattle , Enkephalin, D-Penicillamine (2,5)-/chemistry , Enkephalin, D-Penicillamine (2,5)-/pharmacokinetics , Mice , Octanols/metabolism , Permeability , Sodium Chloride/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...