Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Drug Metab Dispos ; 48(8): 622-630, 2020 08.
Article in English | MEDLINE | ID: mdl-32482756

ABSTRACT

Organic anion transporting polypeptide (OATP) 1B3 is a drug transporter expressed at the basolateral membrane of human hepatocytes. Along with other transporters, including OATP1B1, Na+/taurocholate cotransporting polypeptide (NTCP), and organic cation transporter (OCT) 1, it is responsible for the uptake of endo- and xenobiotics into hepatocytes. Our previous studies demonstrated that OATP1B3 can form hetero-oligomers with OATP1B1 in human embryonic kidney 293T (HEK293) cells and with NTCP in both HEK293 cells and frozen human liver sections. To further characterize the hetero-oligomerization of OATP1B3, we investigated OCT1 as a potential interacting partner and determined the functional consequences of OATP1B3 hetero-oligomerization. We demonstrated interactions between OATP1B3 and OCT1 by coimmunoprecipitation with an anti-OATP1B3 antibody from human hepatocytes. In addition, we visualized the interaction using the proximity ligation assay in both HEK293 cells and in frozen human liver sections. We investigated the functional consequences of OATP1B3 hetero-oligomerization by measuring the OATP1B3 plasma membrane expression and the uptake of the OATP1B3 selective substrate cholecystokinin-8 (CCK-8) in the absence and presence of OATP1B1, NTCP, and OCT1. A significant decrease of OATP1B3 plasma membrane expression was observed after coexpression with OCT1, whereas coexpression with OATP1B1 or NTCP resulted in an increase of plasma membrane expression. With respect to transport, coexpression of OCT1 increased the apparent turnover rate of OATP1B3, whereas coexpression of OATP1B1 or NTCP decreased it. These findings demonstrated that coexpression of OATP1B3 with OATP1B1, NTCP, and OCT1 in HEK293 cells results in a transporter-dependent modification of OATP1B3-mediated CCK-8 transport and suggest that functional results obtained in single transporter overexpressing cell lines over- or underestimate OATP1B3 function in human hepatocytes. SIGNIFICANCE STATEMENT: Coexpression of organic anion transporting polypeptide (OATP) 1B3 with organic cation transporter (OCT) 1, Na+/taurocholate cotransporting polypeptide, or OATP1B1 in human embryonic kidney 293T cells affects its expression level and function. When OCT1 is knocked down in human hepatocytes, function of OATP1B3 goes up. These results suggest that protein-protein interactions can affect the expression and function of the involved proteins, and thus single transporter expression systems might lead to over- or underestimation of drug-drug interactions.


Subject(s)
Liver-Specific Organic Anion Transporter 1/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Cation Transporter 1/metabolism , Sincalide/pharmacokinetics , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Symporters/metabolism , Adult , Cells, Cultured , Child , Gene Expression Regulation , HEK293 Cells , Hepatocytes , Humans , Male , Primary Cell Culture , Protein Multimerization
2.
Drug Metab Dispos ; 48(9): 830-839, 2020 09.
Article in English | MEDLINE | ID: mdl-32561593

ABSTRACT

Dexamethasone (Dex), a widely prescribed anti-inflammatory drug, was reported to induce liver enlargement (hepatomegaly) in clinical practice and in animal models. However, the underlying mechanisms are not elucidated. Dex is a known activator of pregnane X receptor (PXR). Yes-associated protein (YAP) has been implicated in chemically induced liver enlargement. Here, the roles of PXR and YAP pathways were investigated in Dex-induced hepatomegaly. Upregulation of PXR downstream proteins, including cytochrome P450 (CYP) 3A11, 2B10, and organic anion transporter polypeptide 2 (OATP2), indicated PXR signaling was activated after high dose of Dex (50 mg/kg, i.p.), and Dex at 100 µM activated PXR in the dual-luciferase reporter gene assay. Dex also increased the expression of total YAP, nuclear YAP, and YAP downstream proteins, including connective tissue growth factor and cysteine-rich angiogenic inducer 61, indicating activation of the YAP pathway. Furthermore, nuclear translocation of YAP was promoted by activation of PXR. However, hepatocyte proliferation was inhibited with significant decrease in the expression of proliferation-related proteins cyclin D1 and proliferating cell nuclear antigen as well as other regulatory factors, such as forkhead box protein M1, c-MYC, and epidermal growth factor receptor. The inhibitory effect of Dex on hepatocyte proliferation was likely due to its anti-inflammation effect of suppression of inflammation factors. ß-catenin staining revealed enlarged hepatocytes, which were mostly attributable to the accumulation of lipids, such as triglycerides. In summary, high-dose Dex increased liver size accompanied by enlarged hepatocytes, and this was due to the activation of PXR/YAP and their effects on lipid accumulation but not hepatocyte proliferation. These findings provide new insights for understanding the mechanism of Dex-induced hepatomegaly. SIGNIFICANCE STATEMENT: This study identified the roles of pregnane X receptor (PXR) and yes-associated protein (YAP) pathways in dexamethasone (Dex)-induced hepatomegaly. Dex induced PXR/YAP activation, enlarged hepatocytes, and promoted liver enlargement with lipid accumulation, such as triglycerides. However, hepatocyte proliferation was inhibited by the anti-inflammatory effect of Dex. These findings provide new insights for understanding the mechanism of Dex-induced hepatomegaly.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dexamethasone/adverse effects , Hepatomegaly/chemically induced , Pregnane X Receptor/metabolism , Transcription Factors/metabolism , Animals , Cell Proliferation/drug effects , Disease Models, Animal , HEK293 Cells , Hep G2 Cells , Hepatocytes , Hepatomegaly/pathology , Humans , Lipid Metabolism/drug effects , Liver/drug effects , Liver/pathology , Male , Mice , Sincalide/pharmacokinetics , Triglycerides/analysis , Triglycerides/metabolism , YAP-Signaling Proteins
3.
J Med Chem ; 62(3): 1407-1419, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30624060

ABSTRACT

A group of peptide-based, long-acting, stable, highly selective cholecystokinin 1 receptor (CCK-1R) agonists with the potential to treat obesity has been identified and characterized, based on systematic investigation of synthetic CCK-8 analogues with N-terminal linkage to fatty acids. Sulfated Tyr in such compounds was stable in neutral buffer. CCK-1R selectivity was achieved mostly by introducing d- N-methyl-Asp instead of Asp at the penultimate position of CCK-8. Our compound 9 (NN9056) showed similar in vitro CCK-1R potency and CCK-1R affinity as CCK-8, very high selectivity for CCK-1R over the cholecystokinin 2 receptor (CCK-2R), strong reduction of food intake in lean pigs for up to 48 h after one subcutaneous injection without adverse effects, a plasma half-life of 113 h in minipigs after intravenous injection, and acceptable chemical stability in a neutral liquid formulation. In addition, we found a highly selective CCK-2R agonist by replacing Gly in a CCK-8 derivative with Glu.


Subject(s)
Anti-Obesity Agents/therapeutic use , Obesity/drug therapy , Receptors, Cholecystokinin/agonists , Sincalide/analogs & derivatives , Sincalide/therapeutic use , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacokinetics , Female , Humans , Molecular Structure , Sincalide/pharmacokinetics , Structure-Activity Relationship , Swine
4.
Article in English | MEDLINE | ID: mdl-22227312

ABSTRACT

The anorexigenic effect of cholecystokinin (CCK) is well documented in mammals, but documentation in neonatal chicks is limited. Thus, the present study investigated the mechanism underlying the anorexigenic effect of CCK in neonatal chicks. Intraperitoneal (IP) injection of sulfated CCK(26-33) (CCK8S) significantly decreased food intake in chicks at 60 and 300 nmol/kg. Non-sulfated CCK(26-33) (CCK8) also significantly decreased food intake, but its anorexigenic effect was observed only at the highest dose (300 nmol/kg) and short-lived. However, CCK(30-33) (CCK4) had no effect on food intake. Also, the intracerebroventricular (ICV) injection of CCK8S (0.2 and 1 nmol) significantly decreased food intake in chicks. Similar to IP administration, the anorexigenic effect of CCK8 was weak and CCK4 did not affect food intake. IP and ICV injections of CCK8S caused conditioned aversion and increased plasma corticosterone concentrations, suggesting that their anorexigenic effects might be related to stress and/or malaise. This might be true in ICV-injected CCK8S because co-injection of astressin, a corticotropin-releasing hormone receptor antagonist, tended to attenuate the effect of CCK8S. The present study revealed that N-terminal amino acids and the sulfation of Tyr are important for the anorexigenic effect of CCK8S after IP and ICV administered in chicks. Additionally, the effect of central CCK8S might be related to stress and/or malaise.


Subject(s)
Appetite Depressants/pharmacology , Chickens/physiology , Eating/drug effects , Feeding Behavior/drug effects , Sincalide/analogs & derivatives , Animals , Animals, Newborn , Appetite Depressants/administration & dosage , Avoidance Learning/drug effects , Conditioning, Psychological/drug effects , Corticosterone/blood , Corticotropin-Releasing Hormone/pharmacology , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Injections, Intraventricular , Locomotion/drug effects , Male , Peptide Fragments/pharmacology , Photic Stimulation , Sincalide/administration & dosage , Sincalide/pharmacokinetics , Tetragastrin/pharmacology , Time Factors
6.
J Pharmacol Exp Ther ; 311(1): 139-46, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15159445

ABSTRACT

Pitavastatin, a novel potent 3-hydroxymethylglutaryl-CoA reductase inhibitor, is selectively distributed to the liver in rats. However, the hepatic uptake mechanism of pitavastatin has not been clarified yet. In the present study, we investigated the contribution of organic anion transporting polypeptide 2 (OATP2/OATP1B1) and OATP8 (OATP1B3) to pitavastatin uptake using transporter-expressing HEK293 cells and human cryopreserved hepatocytes. Uptake studies using OATP2- and OATP8-expressing cells revealed a saturable and Na(+)-independent uptake, with K(m) values of 3.0 and 3.3 microM for OATP2 and OATP8, respectively. To determine which transporter is more important for its hepatic uptake, we proposed a methodology for estimating their quantitative contribution to the overall hepatic uptake by comparing the uptake clearance of pitavastatin with that of reference compounds (a selective substrate for OATP2 (estrone-3-sulfate) and OATP8 (cholecystokinin octapeptide) in expression systems and human hepatocytes. The concept of this method is similar to the so-called relative activity factor method often used in estimating the contribution of each cytochrome P450 isoform to the overall metabolism. Applying this method to pitavastatin, the observed uptake clearance in human hepatocytes could be almost completely accounted for by OATP2 and OATP8, and about 90% of the total hepatic clearance could be accounted for by OATP2. This result was also supported by estimating the relative expression level of each transporter in expression systems and hepatocytes by Western blot analysis. These results suggest that OATP2 is the most important transporter for the hepatic uptake of pitavastatin in humans.


Subject(s)
Estrone/analogs & derivatives , Liver-Specific Organic Anion Transporter 1/metabolism , Liver/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Quinolines/pharmacokinetics , Biological Transport , Blotting, Western , Cells, Cultured , Drug Interactions , Estrone/pharmacokinetics , Hepatocytes/metabolism , Humans , Sincalide/pharmacokinetics , Sodium/pharmacology , Solute Carrier Organic Anion Transporter Family Member 1B3
7.
Cancer Biother Radiopharm ; 19(1): 93-8, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15068617

ABSTRACT

Cholecystokinin (CCK) receptors of the subtype B (CCK-BR) have been shown to be overexpressed in certain neuroendocrine tumors including medullary thyroid cancer. Our recent work has focused on new methods to radiolabel the CCK8 peptide with 111In or 99mTc for CCK-B receptor imaging. Derivatives of CCK8 were obtained by addition at the N-terminus in solid phase of a DTPA derivative (DTPAGlu) linked through a glycine spacer (DTPAGlu-G-CCK8) or cysteine, glycine and a diphenylphosphinopropionyl moiety (PhosGC-CCK8) for labeling with 111In and 99mTc, respectively. CCK-BR overexpressing A431 cancer cell lines were utilized to characterize in vitro properties of the two compounds as well as for generating xenografts in nude mice for in vivo characterization. Both 111In-DTPAGlu-G-CCK8 and 99mTcPhosGC-CCK8 showed similar binding affinities for CCK-BR with dissociation constants of 20-40 nM, were internalized after interaction with the receptor and displayed prolonged cellular retention times. Specific in vivo interaction with the receptor of both CCK8 analogs was observed in our animal model. 111In-DTPAGlu-G-CCK8 showed better target to non-target ratios, although it appeared to be rapidly metabolized after injection and activity cleared through the kidneys. 99mTc-PhosGC-CCK8 was more stable in vivo but showed marked hepatobiliary clearance with resulting high background activity in the bowel. The rapid clearance and lower background obtained with 111In-DTPAGlu-G-CCK8 make this a better candidate for further development.


Subject(s)
Indium Radioisotopes/metabolism , Radionuclide Imaging/methods , Radiopharmaceuticals , Receptor, Cholecystokinin B/metabolism , Sincalide/metabolism , Technetium/metabolism , Animals , Cell Line, Tumor , Chromatography, High Pressure Liquid , Drug Stability , Humans , Indium Radioisotopes/chemistry , Indium Radioisotopes/pharmacokinetics , Mice , Mice, Nude , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Receptor, Cholecystokinin B/analysis , Sincalide/pharmacokinetics , Technetium/chemistry , Technetium/pharmacokinetics , Tissue Distribution
8.
J Nucl Med ; 45(3): 485-94, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15001692

ABSTRACT

UNLABELLED: Regulatory peptides and their analogs are being extensively investigated as radiopharmaceuticals for cancer imaging and therapy. Receptors of the cholecystokinin family have been shown to be overexpressed in different types of neuroendocrine tumors. The purposes of this study were to evaluate the cholecystokinin octapeptide amide (CCK8) peptide tagged with a diethylenetriaminepentaacetic acid derivative (DTPAGlu) and to test whether a (111)In-labeled conjugate ((111)In-DTPAGlu-G-CCK8, a derivative containing the chelating agent DTPAGlu bound through a glycine linker at the N-terminal end of the bioactive peptide CCK8) is suitable for cholecystokinin-B receptor (CCKBR) imaging. METHODS: CCK8 was synthesized by solid-phase techniques and covalently coupled to DTPAGlu through a glycine linker at its amino terminus. The compound was labeled with (111)In. The radiochemical purity and stability of the compound were assessed by chromatographic methods. NIH-3T3 and A431 cells overexpressing CCKBR were used to characterize the in vitro properties of the compound. Nude mice bearing control and CCKBR-overexpressing A431 xenografts were used as an in vivo model. RESULTS: DTPAGlu-G-CCK8 showed rapid and efficient labeling with (111)In. The radiolabeled conjugate showed specific binding to both cell lines overexpressing CCKBR. Binding was saturable, with a dissociation constant of approximately 20 nmol/L in both cell systems. Both cell lines showed internalization of the ligand after interaction with the receptor. Biodistribution studies showed rapid localization of (111)In-DTPAGlu-G-CCK8 on CCKBR-overexpressing A431 xenografts that was severalfold higher than that on control tumors at all time points tested. Unbound activity showed rapid clearance of over 80% through the kidneys by 30 min after injection. The labeled peptide conjugate was very stable in serum but showed a rapid breakdown after injection. Incubation with kidney homogenates suggested that most breakdown occurred in the kidneys, favoring the clearance of unbound activity. CONCLUSION: Our findings indicate that the in vitro and in vivo characteristics of (111)In-DTPAGlu-G-CCK8 are favorable for CCKBR imaging, as the peptide shows high-affinity binding to the receptor, is internalized in CCKBR-expressing cells, and shows avid uptake in CCKBR-overexpressing xenografts, with rapid clearance of unbound radioactivity through the kidneys. Furthermore, the ease of synthesis, high labeling efficiency, and chemical stability of DTPAGlu make this chelating moiety an ideal candidate for widespread use in peptide radiolabeling for nuclear medicine applications.


Subject(s)
Carcinoma, Squamous Cell/diagnostic imaging , Carcinoma, Squamous Cell/metabolism , Pentetic Acid/pharmacokinetics , Receptor, Cholecystokinin B/metabolism , Sincalide/pharmacokinetics , 3T3 Cells , Animals , Cell Line, Tumor , Humans , Metabolic Clearance Rate , Mice , Mice, Nude , Pentetic Acid/analogs & derivatives , Radiopharmaceuticals/pharmacokinetics , Sincalide/analogs & derivatives , Tissue Distribution , Tomography, Emission-Computed/methods
9.
J Neurochem ; 85(2): 454-61, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12675921

ABSTRACT

A heterogeneity of CCK2 receptors has been reported which could correspond to different states of coupling to G proteins and/or association with different second messenger systems. To investigate these hypotheses, the wild-type CCK2 receptor and three mutants F347A, D100N and K333M/K334T/R335L, expected to modify the coupling of the G protein with the third intracellular loop of the receptor, were transfected into Cos-7 cells and their binding and signalling properties were evaluated using the natural ligand CCK8. Activation of wild-type as well as F347A, D100N or K333M/K334T/R335L CCK2 receptors by this ligand led to a similar arachidonic acid release which was blocked by pertussis toxin and the phospholipase A2 inhibitor, mepacrine. Nevertheless, in contrast to the wild-type CCK2 receptor, addition of CCK8 to cells transfected with the F347A or K333M/K334T/R335L mutants did not result in the production of inositol phosphates while the maximum increase in this second messenger formation was reduced by 30% with the D100N mutant. Taken together, these results suggest that the CCK2 receptor is coupled to two G proteins and that Phe347 and the cluster of basic residues K333/K334/R335 probably play a key role in Gq protein stimulation leading to inositol phosphate production but not in activation of the G protein coupled to phospholipase A2. These data bring additional support at the molecular level to the existence of different affinity states of CCK2 receptors suggested from the results of binding assays and behavioural studies.


Subject(s)
Receptors, Cholecystokinin/metabolism , Signal Transduction/physiology , Amino Acid Substitution , Animals , Arachidonic Acid/metabolism , Binding, Competitive , COS Cells , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Inositol Phosphates/biosynthesis , Ligands , Mutagenesis, Site-Directed , Pertussis Toxin/pharmacology , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Radioligand Assay , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/agonists , Receptors, Cholecystokinin/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Sincalide/pharmacokinetics , Transfection , Tritium
10.
Gastroenterology ; 121(5): 1185-90, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11677211

ABSTRACT

BACKGROUND & AIMS: Cholecystokinin (CCK) is a major gastrointestinal peptide hormone that is released postprandially from the small intestine and exerts marked effects on gallbladder and gastrointestinal motility. The smaller isoforms CCK-8 and CCK-4 are rapidly taken up into hepatocytes, metabolized, and excreted into bile. Our aim was to identify and characterize the hepatocellular CCK-8 uptake system. METHODS: CCK-8 uptake was measured in Xenopus laevis oocytes expressing the organic anion-transporting polypeptides of rat liver (Oatp1, Oatp2, Oatp3, or Oatp4) and of human liver (OATP-A, OATP-B, OATP-C, or OATP8) and in primary cultured rat hepatocytes. RESULTS: Rat Oatp4 and human OATP8 efficiently mediated CCK-8 uptake in oocytes, with Michaelis constant (Km) values of 14.9 +/- 2.9 micromol/L and 11.1 +/- 2.9 micromol/L, respectively. CCK-8 uptake by hepatocytes was also saturable, with a Km of 6.7 +/- 2.1 micromol/L. The Km value in rat hepatocytes is consistent with Oatp4-mediated transport. CONCLUSIONS: CCK-8 is selectively transported by rat Oatp4 and human OATP8, both of which are exclusively expressed at the basolateral membrane of hepatocytes. These 2 transporters are the first and probably the predominant hepatic uptake systems for CCK-8 and may be critical for the rapid clearance of this hormone from the circulation.


Subject(s)
Liver/metabolism , Organic Anion Transporters, Sodium-Independent/physiology , Organic Anion Transporters/physiology , Sincalide/pharmacokinetics , Animals , Cells, Cultured , Hepatocytes/metabolism , Humans , Male , Rats , Rats, Sprague-Dawley , Solute Carrier Organic Anion Transporter Family Member 1B3 , Xenopus laevis
11.
Brain Res ; 860(1-2): 11-20, 2000 Mar 31.
Article in English | MEDLINE | ID: mdl-10727619

ABSTRACT

The aim of the study was to determine whether paraventricular nucleus (PVN) injection of an anorexic 500-pmol dose of cholecystokinin (CCK)-8 could increase plasma CCK-8 levels sufficiently to suppress feeding by a peripheral mechanism. Rats received PVN injections of CCK-8 either alone or with 3H-labelled propionylated CCK-8 (3H-pCCK-8) and plasma samples were taken at various times from 3 to 120 min post-injection. Plasma CCK-8 levels were estimated from measurements of both total plasma CCK-like immunoreactivity (CCK-LI) and 3H-pCCK-8 activity. PVN injections of CCK-8 and 3H-pCCK-8 produced estimated peak increases in plasma CCK-8 of 15+/-11 and 22+/-3 pM, respectively. The i.v. infusion of CCK-8 doses (0.2 and 1 nmol/kg h) that bracketed the threshold dose for suppression of feeding, increased plasma CCK-LI from a basal level of 6+/-1 to 49+/-10 and 166+/-36 pM, respectively. The i.v. injections of 600 and 4800 pmol of CCK-8 did not suppress feeding. These results suggest that PVN injection of an anorexic 500-pmol dose of CCK-8 does not increase plasma CCK-8 levels sufficiently to suppress feeding by a peripheral mechanism.


Subject(s)
Appetite Depressants/pharmacokinetics , Paraventricular Hypothalamic Nucleus/drug effects , Sincalide/pharmacokinetics , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/analysis , Eating/drug effects , Infusions, Intravenous , Injections , Injections, Intravenous , Male , Paraventricular Hypothalamic Nucleus/physiology , Rats , Rats, Sprague-Dawley , Sincalide/administration & dosage , Sincalide/blood
12.
Pharmacol Biochem Behav ; 59(2): 439-44, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9476993

ABSTRACT

Cholecystokinin octapeptide (CCK-8) and the peptide analog ARL 14294, formerly FPL 14294, [Hpa(SO3H)-Met-Gly-Trp-Met-Asp-N(Me)Phe-NH2], have been reported to induce satiety by interaction with the CCK-A receptor subtype. ARL 15849 [Hpa(SO3H)-Nle-Gly-Trp-Nle-N(Me)-Asp-Phe-NH2] is an improved ARL 14294 analog with enhanced CCK-A receptor selectivity, greater stability, and a longer duration of action. The affinity of ARL 15849 for the CCK-A receptor (Ki = 0.034 nM) is 6,600 fold greater than for the CCK-B receptor (Ki = 224 nM), whereas CCK-8 and ARL 14294 are nonselective. Although comparable in potency to contract isolated gallbladder and induce pancreatic phosphatidylinositol hydrolysis, ARL 15849 is 3- and 100-fold more potent than ARL 14294 and CCK-8, respectively, to inhibit 3-h feeding in rats. The duration of feeding inhibition was significantly longer for ARL 15849 (>5 h), compared to equipotent doses of ARL 14294 (3 h), and CCK-8 (1 h). Intranasal administration of ARL 15849 inhibits feeding in beagle dogs with a greater separation between doses that induce emesis and those that inhibit feeding. Therefore, ARL 15849 is a potent, selective, intranasally active anorectic agent which may be useful in the treatment of eating disorders.


Subject(s)
Appetite Depressants/pharmacology , Receptors, Cholecystokinin/agonists , Sincalide/analogs & derivatives , Animals , Appetite Depressants/pharmacokinetics , Dogs , Dose-Response Relationship, Drug , Eating/drug effects , Gallbladder/drug effects , Gallbladder/metabolism , Guinea Pigs , In Vitro Techniques , Injections, Intraperitoneal , Pancreas/drug effects , Pancreas/metabolism , Phosphatidylinositols/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A , Sincalide/pharmacokinetics , Sincalide/pharmacology
13.
Regul Pept ; 55(2): 179-88, 1995 Jan 26.
Article in English | MEDLINE | ID: mdl-7754104

ABSTRACT

Rats were given i.v., intranasal or intraperitoneal doses of CCK-8 (sulfated) labelled with 125I-labeled Bolton and Hunter reagent. Radioactivity was found mainly in the liver, kidney, and the intestinal contents. No radioactivity was detected in the brain. In animals dosed i.v., specific localization occurred in the tissue of the pyloric region of the stomach, and in the pancreas. Label persisted within the pyloric region of the stomach for longer than 30 min, in spite of the reported half-life of CCK-8 in plasma of approximately 1 min. Intranasal and intraperitoneal doses had limited bioavailability. The binding to the sites in the pyloric region of the stomach, which required systemic delivery, may have identified receptors associated with appetite control.


Subject(s)
Sincalide/analysis , Administration, Intranasal , Amino Acid Sequence , Animals , Autoradiography , Biological Availability , Brain/metabolism , Female , Frozen Sections , Injections, Intraperitoneal , Injections, Intravenous , Intestinal Mucosa/metabolism , Intestines/anatomy & histology , Iodine Isotopes , Isotope Labeling , Kidney/metabolism , Liver/metabolism , Molecular Sequence Data , Pancreas/anatomy & histology , Pancreas/metabolism , Pylorus/anatomy & histology , Pylorus/metabolism , Rats , Rats, Inbred F344 , Sincalide/administration & dosage , Sincalide/chemistry , Sincalide/pharmacokinetics , Tissue Distribution
14.
Am J Physiol ; 267(5 Pt 2): R1309-19, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7977859

ABSTRACT

Methods developed previously for studying the effect of cerebroventricular injection or ventriculocisternal perfusion of test substances are unsatisfactory because the test substance is not confined to the central compartment. Most likely the test substance enters the peripheral circulation via the arachnoid villi. The purpose of this paper is to describe a method for perfusing the cerebroventricular system of conscious dogs without passage of test substances to the peripheral circulation. With the method described, the mean (+/- SE) cerebroventricular pressure in conscious dogs was 7.4 +/- 0.8 cmH2O (n = 16), and the mean (+/- SE) production of cerebrospinal fluid (CSF) was 25 +/- 0.3 microliter/min (n = 16). Endogenously occurring migrating myoelectric complexes (MMCs) of the small intestine were recorded in dogs before catheters were implanted in the left and right lateral ventricles and the fourth ventricle and after catheter implantation during cerebroventricular perfusion with artificial CSF alone or with CSF containing sulfated (S-CCK-OP) or nonsulfated cholecystokinin octapeptide (NS-CCK-OP). Only cerebroventricular perfusion with S-CCK-OP (1.2 pmol.kg-1.min-1; n = 20) replaced spontaneously occurring MMCs with a fed-like pattern of myoelectric activity. The results suggest that replacement of the fasting pattern of myoelectric activity with a fed-like pattern in the fasted dog was mediated by CCK-A receptors located in one or more brain nuclei surrounding the third ventricle.


Subject(s)
Cerebral Ventricles/physiology , Myoelectric Complex, Migrating/drug effects , Sincalide/analogs & derivatives , Sincalide/pharmacology , Sincalide/pharmacokinetics , Animals , Blood Circulation , Catheters, Indwelling , Cerebral Ventricles/drug effects , Dogs , Female , Infusions, Intravenous , Iodine Radioisotopes , Perfusion/methods , Radioisotope Dilution Technique , Sincalide/administration & dosage , Tissue Distribution
15.
Alcohol Alcohol ; 29(5): 575-81, 1994 Sep.
Article in English | MEDLINE | ID: mdl-7811341

ABSTRACT

Brain cholecystokinin (CCK) receptors have been implicated in anxiety disorders and suicidal behaviour. We have examined the radioligand binding ability of CCK and benzodiazepine receptors in rat brain after long-term intermittent voluntary vs voluntary and forced low-dose ethanol exposure. During 58 weeks, one group of rats had a choice between ethanol and water as the drinking fluid for 24 hr each week. Another group of rats had the same weekly choice between ethanol and water, but at the end of each 24 hr choice period, ethanol (2.0 g/kg) was injected. During the second period of ethanol treatment, lasting for 32 weeks, both ethanol-treated groups had continuous free access to ethanol and water. These two treatments have previously been shown to induce partially different neurochemical alterations. In the present investigation, benzodiazepine receptor binding in the frontal cortex, hippocampus and striatum was similar in both ethanol treatment groups compared to controls. CCK receptor binding in the hippocampus and striatum did not differ between the three groups; however, in the frontal cortex, there was an increase in the apparent number of CCK binding sites in the group of rats submitted to voluntary plus forced ethanol exposure as compared to the control group or the voluntary intake group. These results suggest that long-term ethanol treatment may lead to alterations in brain CCK-ergic neurotransmission, but that the changes are specific to the treatment schedule.


Subject(s)
Alcoholism/physiopathology , Brain/physiopathology , Receptors, Cholecystokinin/physiology , Animals , Binding, Competitive , Brain/drug effects , Corpus Striatum/drug effects , Corpus Striatum/physiopathology , Dose-Response Relationship, Drug , Flunitrazepam/pharmacokinetics , Frontal Lobe/drug effects , Frontal Lobe/physiopathology , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Cholecystokinin/drug effects , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Sincalide/pharmacokinetics
16.
Pharmacol Biochem Behav ; 47(3): 701-8, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8208791

ABSTRACT

Cholecystokinin octapeptide (CCK-8) induces satiety in many species including man. However, its therapeutic utility is restricted due to its short biological half-life and poor bioavailability. FPL 14294 [4-(sulfoxy)-phenylacetyl(MePhe6)CCK-6] is a CCK analog with enhanced metabolic stability that was comparable to CCK-8 in potency to contract isolated gallbladder and in affinity at the CCK-A and CCK-B receptor. However, FPL 14294 was more than 200 times more potent than CCK-8 in inhibiting 3-h feeding in 21-h fasted rats. FPL 14294 also possessed intranasal anorectic activity at 5 micrograms/kg, while CCK-8 was inactive at doses up to 500 micrograms/kg. Anorectic activity was inhibited by pretreatment with a CCK-A antagonist (MK-329) but not by a CCK-B antagonist (L365,260). The anorectic effects of CCK-8 and FPL 14294 were the result of a direct effect on feeding and not caused indirectly by effects on water intake. These results indicate that FPL 14294 is a potent, intranasally active, anorectic agent whose enhanced in vivo potency over that of CCK-8 may reflect differences in stability, bioavailability, or receptor kinetics.


Subject(s)
Appetite Depressants/pharmacology , Sincalide/analogs & derivatives , Sincalide/physiology , Administration, Intranasal , Amino Acid Sequence , Animals , Appetite Depressants/administration & dosage , Appetite Depressants/pharmacokinetics , Biological Availability , Dogs , Drinking/drug effects , Gallbladder/drug effects , Guinea Pigs , Half-Life , In Vitro Techniques , Kidney Cortex/metabolism , Male , Molecular Sequence Data , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Pancreas/drug effects , Pancreas/metabolism , Rabbits , Rats , Rats, Sprague-Dawley , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/drug effects , Receptors, Cholecystokinin/metabolism , Sincalide/administration & dosage , Sincalide/pharmacokinetics , Sincalide/pharmacology
17.
Gastroenterology ; 105(6): 1732-6, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8253350

ABSTRACT

BACKGROUND: Cholecystokinin (CCK) 58 is the predominant molecular form of CCK in canine and human intestine and circulating blood. There is no report on the metabolism and clearance rate of CCK-58. The aim of this study was to compare the in vivo half-life and metabolism of CCK-58 with that of synthetic CCK-8. METHODS: CCK-58 was purified from canine intestine by consecutive high-performance liquid chromatographic (HPLC) and fast protein liquid chromatographic steps. The peptides were given to 12 dogs as an intravenous (IV) bolus injection to determine the half-life of circulating CCK. Six dogs were given CCK-58 or CCK-8 as a constant IV infusion to determine plasma clearance rates and stability in circulating blood. Circulating molecular forms of CCK were determined by radioimmunoassay after extraction of CCK from plasma and characterization by HPLC. RESULTS: The half-life of CCK-58 was 4.4 +/- 0.6 minutes compared with 1.3 +/- 0.1 minutes for CCK-8. Less than 5% of CCK-58 could be detected as smaller forms during constant IV infusion. CONCLUSIONS: The longer half-life of CCK-58 compared with CCK-8 and the minimal conversion into smaller forms during constant IV infusion are consistent with the finding that CCK-58 is not only the major stored form but also the circulating form of CCK after endogenous stimulation in dogs.


Subject(s)
Cholecystokinin/pharmacokinetics , Sincalide/pharmacokinetics , Animals , Cholecystokinin/administration & dosage , Cholecystokinin/isolation & purification , Dogs , Half-Life , Infusions, Intravenous , Injections, Intravenous , Intestinal Mucosa/chemistry , Metabolic Clearance Rate , Sincalide/administration & dosage , Sincalide/chemical synthesis
18.
Gastroenterol Nurs ; 14(2): 98-100, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1932168

ABSTRACT

Sincalide is a rapid-acting, synthetic analog of cholecystokinin for intravenous use in postevacuation cholecystography. Serious reactions to sincalide have not been reported. The intravenous administration of sincalide causes a prompt contraction of the gallbladder as compared to the stimulus of a fatty meal which causes progressive contraction that becomes maximal in about 40 minutes. The use of Kinevac to accelerate the transit time through the small bowel decreases the time and extent of radiation associated with fluoroscopy and length of the x-ray examination of the intestinal tract. Duodenal aspiration obtained after the administration of Kinevac provides a sample of concentrated bile for analysis of cholesterol, bile salts, phospholipids and crystals. When used in conjunction with secretin to stimulate pancreatic secretion, an aspirate is readily obtained for analysis of enzyme activity, composition and cytology. As the development of endoscopic manometry affords a modality to measure and record sphincter of Oddi pressures, the paradoxical responses noted to the intravenous administration of CCK during manometric evaluation supports the diagnostic value of Kinevac used as a provocative agent in the evaluation of biliary dyskinesia.


Subject(s)
Cholecystography/methods , Sincalide/therapeutic use , Humans , Sincalide/administration & dosage , Sincalide/pharmacokinetics
19.
J Pharmacol Exp Ther ; 257(3): 1076-80, 1991 Jun.
Article in English | MEDLINE | ID: mdl-2046021

ABSTRACT

Agonists and antagonists selective for the brain-type [cholecystokinin (CCK)-B] and the peripheral-type (CCK-A) CCK receptor were used to localize the site(s) of action at which CCK inhibits food consumption. BC 264, a highly selective CCK-B receptor agonist, did not decrease consumption of a palatable meal when administered either i.p. or into the lateral ventricles of the brain, whereas CCK decreased feeding when administered i.p. at the same doses. CCK decreased feeding when administered i.v.t. at a high dose, 5 micrograms. L-364,718, an antagonist selective for the CCK-A receptor, blocked completely the action of centrally administered CCK, whereas L-365,260, a selective CCK-B receptor antagonist, had no effect on the ability of centrally administered CCK to inhibit feeding. To estimate the quantity of i.v.t. administered CCK which reached the periphery, a tracer of radiolabeled [3H]p-CCK8 ([3H]CCK octapeptide sulfate), combined with unlabeled pCCK8 (5 micrograms) was administered i.c.t. Thirty minutes after administration, intact radiolabeled pCCK8 was extracted from the plasma and measured in the blood in nanomolar concentrations, exceeding the amounts of CCK octapeptide sulfate reported previously to be present in the plasma after a meal. Intraventricularly administered CCK thus appears to reduce feeding in the rat through a mechanism involving a CCK-A receptor subtype in the periphery.


Subject(s)
Cholecystokinin/pharmacology , Eating/drug effects , Phenylurea Compounds , Receptors, Cholecystokinin/physiology , Animals , Benzodiazepinones/pharmacology , Cholecystokinin/analogs & derivatives , Cholecystokinin/antagonists & inhibitors , Devazepide , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Injections, Intraventricular , Male , Peptide Fragments/pharmacology , Rats , Rats, Inbred Strains , Receptors, Cholecystokinin/drug effects , Sincalide/analogs & derivatives , Sincalide/pharmacokinetics , Sincalide/pharmacology
20.
Am J Physiol ; 260(6 Pt 1): G815-9, 1991 Jun.
Article in English | MEDLINE | ID: mdl-2058671

ABSTRACT

We have described previously that the gallbladder responds abnormally to infusions of cholecystokinin octapeptide (CCK-8) in patients with irritable bowel syndrome (IBS). To confirm these results and to examine the possible mechanisms, patients with IBS and predominant symptoms of diarrhea or constipation were compared with matched controls. During infusions of CCK-8 at one of three doses, the response of the gallbladder was measured ultrasonographically. The levels of CCK-8 reached in the peripheral circulation and degradation of the peptide in vitro and in vivo were used to evaluate metabolism of cholecystokinin. We confirmed that the gallbladders of patients with IBS responded abnormally to CCK-8; however, the differences were not due to any prereceptor event. Instead, this abnormality in IBS must be explained by an atypical response at the level of the target tissues.


Subject(s)
Colonic Diseases, Functional/physiopathology , Gallbladder/physiopathology , Muscle, Smooth/physiopathology , Sincalide/pharmacology , Adult , Colonic Diseases, Functional/classification , Colonic Diseases, Functional/diagnostic imaging , Female , Gallbladder/diagnostic imaging , Gallbladder/drug effects , Gallbladder/physiology , Humans , Kinetics , Male , Muscle, Smooth/diagnostic imaging , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Reference Values , Sincalide/pharmacokinetics , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL
...