Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Appetite ; 127: 334-340, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29782892

ABSTRACT

Combination approaches for the treatment of metabolic diseases such as obesity and diabetes are becoming increasingly relevant. Co-administration of a glucagon-like peptide-1 receptor (GLP-1R) agonist with a cholecystokinin receptor-1 (CCKR1) agonist exert synergistic effects on weight loss in obese rodents. Here, we report on the effects of a novel fusion peptide (C2816) comprised of a stabilized GLP-1R agonist, AC3174, and a CCKR1-selective agonist, AC170222. C2816 was constructed such that AC3174 was linked to the N-terminus of AC170222, thus preserving the C-terminal amide of the CCK moiety. In functional in vitro assays C2816 retained full agonism at GLP-1R and CCKR1 at lower potency compared to parent molecules, whereas a previously reported fusion peptide in the opposite orientation, (pGlu-Gln)-CCK-8/exendin-4, exhibited no activity at either receptor. Acutely, in vivo, C2816 increased cFos in key central nuclei relevant to feeding behavior, and reduced food intake in wildtype (WT), but less so in GLP-1R-deficient (GLP-1RKO), mice. In sub-chronic studies in diet-induced obese (DIO) mice, C2816 exerted superior reduction in body weight compared to co-administration of AC3174 and AC170222 albeit at a higher molar dose. These data suggest that the synergistic pharmacological effects of GLP-1 and CCK pathways can be harnessed in a single therapeutic peptide.


Subject(s)
Anti-Obesity Agents/chemistry , Cholecystokinin/chemistry , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide-1 Receptor/agonists , Receptor, Cholecystokinin A/agonists , Animals , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/pharmacology , Brain/drug effects , Cholecystokinin/administration & dosage , Drug Synergism , Eating/drug effects , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide-1 Receptor/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/drug therapy , Peptides/administration & dosage , Peptides/chemistry , Peptides/pharmacology , Rats, Sprague-Dawley , Weight Loss
2.
Trends Endocrinol Metab ; 27(9): 609-619, 2016 09.
Article in English | MEDLINE | ID: mdl-27156041

ABSTRACT

Cholecystokinin (CCK) regulates appetite and reduces food intake by activating the type 1 CCK receptor (CCK1R). Attempts to develop CCK1R agonists for obesity have yielded active agents that have not reached clinical practice. Here we discuss why, along with new strategies to target CCK1R more effectively. We examine signaling events and the possibility of developing agents that exhibit ligand-directed bias, to dissociate satiety activity from undesirable side effects. Potential allosteric sites of modulation are also discussed, along with desired properties of a positive allosteric modulator (PAM) without intrinsic agonist action as another strategy to treat obesity. These new types of CCK1R-active drugs could be useful as standalone agents or as part of a rational drug combination for management of obesity.


Subject(s)
Receptors, Cholecystokinin/metabolism , Allosteric Regulation/genetics , Allosteric Regulation/physiology , Animals , Cholecystokinin/metabolism , Humans , Obesity/genetics , Obesity/metabolism , Receptor, Cholecystokinin A/agonists , Receptor, Cholecystokinin A/metabolism , Receptors, Cholecystokinin/agonists , Receptors, Cholecystokinin/genetics
3.
J Med Chem ; 58(24): 9562-77, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-26654202

ABSTRACT

The type 1 cholecystokinin receptor (CCK1R) has multiple physiologic roles relating to nutrient homeostasis, including mediation of postcibal satiety. This effect has been central in efforts to develop agonists of this receptor as part of a program to manage and/or prevent obesity. While a number of small molecule CCK1R agonists have been developed, none have yet been approved for clinical use, based on inadequate efficacy, side effects, or the potential for toxicity. Understanding the molecular details of docking and mechanism of action of these ligands can be helpful in the rational refinement and enhancement of small molecule drug candidates. In the current work, we have defined the mechanism of binding and activity of two triazolobenzodiazepinones, CE-326597 and PF-04756956, which are reported to be full agonist ligands. To achieve this, we utilized receptor binding with a series of allosteric and orthosteric radioligands at structurally related CCK1R and CCK2R, as well as chimeric CCK1R/CCK2R constructs exchanging residues in the allosteric pocket, and assessment of biological activity. These triazolobenzodiazepinones docked within the intramembranous small molecule allosteric ligand pocket, with higher affinity binding to CCK2R than CCK1R, yet with biological activity exclusive to or greatly enhanced at CCK1R. These ligands exhibited cooperativity with benzodiazepine binding across the CCK1R homodimeric complex, resulting in their ability to inhibit only a fraction of the saturable binding of a benzodiazepine radioligand, unlike other small molecule antagonists and agonists of this receptor. This may contribute to the understanding of the unique short duration and reversible gallbladder contraction observed in vivo upon administration of these drugs.


Subject(s)
Benzodiazepines/chemistry , Benzodiazepinones/chemistry , Receptor, Cholecystokinin A/agonists , Triazoles/chemistry , Allosteric Site , Amino Acid Sequence , Animals , Benzodiazepines/pharmacology , Benzodiazepinones/pharmacology , CHO Cells , Cricetulus , Humans , Molecular Docking Simulation , Molecular Sequence Data , Mutation , Protein Multimerization , Radioligand Assay , Rats , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Structure-Activity Relationship , Triazoles/pharmacology
4.
Diabetes Obes Metab ; 17(1): 61-73, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25204356

ABSTRACT

AIM: To test the impact of cholecystokinin (CCK) plus either amylin or a glucagon-like peptide-1 receptor (GLP-1R) agonist on metabolic variables in diet-induced obese (DIO) rodents. METHODS: A stabilized acetylated version of CCK-8 (Ac-Y*-CCK-8), selective CCK1 receptor (CCK1R) or CCK2 receptor (CCK2R) agonists, amylin or the GLP-1R agonist and exenatide analogue AC3174 were administered in select combinations via continuous subcutaneous infusion to DIO rats for 14 days, or Lep(ob) /Lep(ob) mice for 28 days, and metabolic variables were assessed. RESULTS: Combined administration of Ac-Y*-CCK-8 with either amylin or AC3174 induced greater than additive weight loss in DIO rats, with the overall magnitude of effect being greater with AC3174 + Ac-Y*-CCK-8 treatment. Co-infusion of AC3174 with a specific CCK1R agonist, but not a CCK2R agonist, recapitulated the weight loss mediated by AC3174 + Ac-Y*-CCK-8 in DIO rats, suggesting that synergy is mediated by CCK1R activation. In a 4 × 4 full-factorial response surface methodology study in DIO rats, a synergistic interaction between AC3174 and the CCK1R-selective agonist on body weight and food intake was noted. Co-administration of AC3174 and the CCK1R-selective agonist to obese diabetic Lep(ob) /Lep(ob) mice elicited a significantly greater reduction in percentage of glycated haemoglobin and food intake relative to the sum effects of monotherapy groups. CONCLUSIONS: The anti-obesity and antidiabetic potential of combined GLP-1R and CCK1R agonism is an approach that warrants further investigation.


Subject(s)
Anti-Obesity Agents/therapeutic use , Cholecystokinin/analogs & derivatives , Diabetes Mellitus/drug therapy , Hypoglycemic Agents/therapeutic use , Islet Amyloid Polypeptide/therapeutic use , Obesity/drug therapy , Peptides/therapeutic use , Acetylation , Animals , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/adverse effects , Cholecystokinin/administration & dosage , Cholecystokinin/adverse effects , Cholecystokinin/therapeutic use , Diabetes Mellitus/metabolism , Diet, High-Fat/adverse effects , Drug Synergism , Drug Therapy, Combination/adverse effects , Energy Intake/drug effects , Glucagon-Like Peptide-1 Receptor , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Infusions, Subcutaneous , Islet Amyloid Polypeptide/administration & dosage , Islet Amyloid Polypeptide/adverse effects , Male , Mice, Mutant Strains , Obesity/complications , Obesity/etiology , Obesity/metabolism , Peptides/administration & dosage , Peptides/adverse effects , Random Allocation , Rats, Sprague-Dawley , Receptor, Cholecystokinin A/agonists , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin B/agonists , Receptor, Cholecystokinin B/metabolism , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Weight Loss/drug effects
5.
Endocrinology ; 155(5): 1700-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24564397

ABSTRACT

Apolipoprotein AIV (Apo AIV) and cholecystokinin (CCK) are secreted in response to fat consumption, and both cause satiation via CCK 1 receptor (CCK-1R)-containing vagal afferent nerves to the nucleus of the solitary tract (NTS), where Apo AIV is also synthesized. Fasted male Long-Evans rats received ip CCK-8 or fourth-ventricular (i4vt) Apo AIV alone or in combination. Food intake and c-Fos proteins (a product of the c-Fos immediate-early gene) were assessed. i4vt Apo AIV and/or ip CCK at effective doses reduced food intake and activated c-Fos proteins in the NTS and hypothalamic arcuate nucleus and paraventricular nucleus. Blockade of the CCK-1R by i4vt lorglumide adjacent to the NTS attenuated the satiating and c-Fos-stimulating effects of CCK and Apo AIV, alone or in combination. Maintenance on a high-fat diet (HFD) for 10 weeks resulted in weight gain and attenuation of both the behavioral and c-Fos responses to a greater extent than occurred in low-fat diet-fed and pair-fed HFD animals. These observations suggest that NTS Apo AIV or/and peripheral CCK requires vagal CCK-1R signaling to elicit satiation and that maintenance on a HFD reduces the satiating capacity of these 2 signals.


Subject(s)
Apolipoproteins A/metabolism , Appetite Regulation , Cholecystokinin/metabolism , Nerve Tissue Proteins/metabolism , Neurons, Afferent/metabolism , Receptor, Cholecystokinin A/metabolism , Solitary Nucleus/metabolism , Animals , Apolipoproteins A/administration & dosage , Apolipoproteins A/genetics , Apolipoproteins A/pharmacology , Appetite Depressants/administration & dosage , Appetite Depressants/pharmacology , Appetite Depressants/therapeutic use , Appetite Regulation/drug effects , Appetite Stimulants/administration & dosage , Appetite Stimulants/pharmacology , Appetitive Behavior/drug effects , Behavior, Animal/drug effects , Cholecystokinin/administration & dosage , Cholecystokinin/analogs & derivatives , Cholecystokinin/antagonists & inhibitors , Diet, High-Fat/adverse effects , Hormone Antagonists/administration & dosage , Hormone Antagonists/pharmacology , Infusions, Intraventricular , Injections, Intraperitoneal , Male , Nerve Tissue Proteins/administration & dosage , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Neurons, Afferent/drug effects , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Rats , Rats, Long-Evans , Receptor, Cholecystokinin A/agonists , Receptor, Cholecystokinin A/antagonists & inhibitors , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Sincalide/administration & dosage , Sincalide/analogs & derivatives , Sincalide/pharmacology , Solitary Nucleus/drug effects
6.
J Biol Chem ; 288(29): 21082-21095, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23754289

ABSTRACT

Understanding the molecular basis of drug action can facilitate development of more potent and selective drugs. Here, we explore the molecular basis for action of a unique small molecule ligand that is a type 1 cholecystokinin (CCK) receptor agonist and type 2 CCK receptor antagonist, GI181771X. We characterize its binding utilizing structurally related radioiodinated ligands selective for CCK receptor subtypes that utilize the same allosteric ligand-binding pocket, using wild-type receptors and chimeric constructs exchanging the distinct residues lining this pocket. Intracellular calcium assays were performed to determine biological activity. Molecular models for docking small molecule agonists to the type 1 CCK receptor were developed using a ligand-guided refinement approach. The optimal model was distinct from the previous antagonist model for the same receptor and was mechanistically consistent with the current mutagenesis data. This study revealed a key role for Leu(7.39) that was predicted to interact with the isopropyl group in the N1 position of the benzodiazepine that acts as a "trigger" for biological activity. The molecular model was predictive of binding of other small molecule agonists, effectively distinguishing these from 1065 approved drug decoys with an area under curve value of 99%. The model also selectively enriched for agonist compounds, with 130 agonists identified by ROC analysis when seeded in 2175 non-agonist ligands of the type 1 CCK receptor (area under curve 78%). Benzodiazepine agonists in this series docked in consistent pose within this pocket, with a key role played by Leu(7.39), whereas the role of this residue was less clear for chemically distinct agonists.


Subject(s)
Benzodiazepines/pharmacology , Receptor, Cholecystokinin A/agonists , Amino Acid Sequence , Animals , Benzodiazepines/chemistry , CHO Cells , Cricetinae , Cricetulus , Models, Molecular , Molecular Sequence Data , Mutant Proteins/agonists , Mutant Proteins/chemistry , Mutant Proteins/metabolism , ROC Curve , Receptor, Cholecystokinin A/chemistry , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment
7.
J Endocrinol ; 216(1): 53-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23055535

ABSTRACT

Cholecystokinin (CCK) is a hormone that has important physiological effects on energy balance. This study has used a stable CCK(1) receptor agonist, (pGlu-Gln)-CCK-8, to evaluate the metabolic effects of prolonged administration in normal mice. Twice-daily injection of (pGlu-Gln)-CCK-8 for 28 days resulted in significantly lowered body weights (P<0.05) on days 24 and 28, which was associated with decreased accumulated calorie intake (P<0.01) from day 12 onward. Nonfasting plasma glucose was significantly reduced (P<0.05) on day 28, while plasma insulin concentrations were increased (P<0.05). After 28 days, glucose tolerance and glucose-mediated insulin secretion were not significantly different in (pGlu-Gln)-CCK-8-treated mice. However, following a 15-min refeeding period in 18-h fasted mice, glucose levels were significantly (P<0.05) decreased by (pGlu-Gln)-CCK-8 despite similar food intake and nutrient-induced insulin levels. Insulin sensitivity in (pGlu-Gln)-CCK-8-treated mice was significantly (P<0.01) improved compared with controls. Accumulation of triacylglycerol in liver was reduced (P<0.01) but there were no differences in circulating cholesterol and triacylglycerol concentrations, as well as triacylglycerol content of pancreatic, muscle, and adipose tissue in (pGlu-Gln)-CCK-8 mice. These data highlight the beneficial metabolic effects of prolonged (pGlu-Gln)-CCK-8 administration and confirm a lack of detrimental effects.


Subject(s)
Anti-Obesity Agents/pharmacology , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Liver/drug effects , Receptor, Cholecystokinin A/agonists , Signal Transduction/drug effects , Sincalide/analogs & derivatives , Animals , Anti-Obesity Agents/adverse effects , Blood Glucose/analysis , Energy Intake/drug effects , Hyperglycemia/prevention & control , Hypoglycemic Agents/adverse effects , Insulin/blood , Insulin Resistance , Insulin-Secreting Cells/metabolism , Liver/metabolism , Male , Mice , Postprandial Period , Sincalide/adverse effects , Sincalide/pharmacology , Time Factors , Triglycerides/metabolism , Weight Gain/drug effects
8.
Brain Res Bull ; 89(5-6): 177-84, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22981453

ABSTRACT

Cholecystokinin, a neuroactive peptide functioning as a neurotransmitter and neuromodulator in the central nervous system, mediates a number of processes and is implicated in neurological and psychiatric disorders such as Parkinson's disease, anxiety and schizophrenia. Striatum is one of the brain structures with the highest concentrations of CCK in the brain, rich in CCK receptors as well. The physiological effect of CCK on cholinergic interneurons, which are the major interneurons in striatum and the modulatory interactions which exist between dopamine, acetylcholine and cholecystokinin in this brain structure are still unclear. We studied the effect of cholecystokinin octapeptide (CCK-8) on the release of acetylcholine (ACh) from striatal slices of the rat brain. CCK-8 (0.01-0.1µM) showed no statistically significant effect on the basal but enhanced dose-dependently the electrically (2Hz)-evoked release of [(3)H]ACh. When slices were preperfused with 100µM sulpiride, a selective dopamine D(2) receptor antagonist, the CCK-8 (0.01µM) effect on electrically stimulated ACh release was increased nearly 2-fold. A similar increase was observed after depletion of endogenous dopamine (DA) from nigro-striatal dopaminergic neurons with 6-hydroxydopamine (6-OHDA) (2× 250µg/animal, i.c.v.). Furthermore in the presence of dopamine (100µM) or apomorphine (10µM), the prototypical DA receptor agonist, CCK-8 (0.01µM) failed to enhance the stimulation-evoked release of [(3)H]ACh. The D(2) receptor agonist quinpirol (1µM) abolished the CCK-8 effect on electrically stimulated ACh release as well. The increase in electrically induced [(3)H]ACh release produced by 0.01µM CCK-8 was antagonized by d,l loxiglumide (CR 1505), 10µM, a non-peptide CCK-A receptor antagonist and by Suc-Tyr-(OSO3)-Met-Gly-Trp-Met-Asp-ß-phenethyl-amide (GE-410), 1µM, a peptide CCK-A receptor antagonist. The antagonistic effect of GE-410 on the CCK-8-potentiated, electrically induced release of [(3)H]ACh was studied in striatum for the first time. CAM 1028 (10µM), a CCK-B receptor antagonist, also prevented the potentiating effect of CCK-8 (0.01µM) on electrically stimulated release of [(3)H]ACh. The presented results indicate that (i) CCK-8 is capable of increasing ACh elicited by field electrical stimulation in striatum; (ii) CCK-8 is more effective in its ACh-stimulating effect when dopaminergic activity in striatum is blocked i.e. CCK-8-facilitated release of electrically induced ACh from cholinergic interneurons in the striatum is under the inhibitory control of the tonic activity of dopamine from the nigrostriatal pathway; (iii) the enhancing effect of CCK-8 on electrically evoked ACh release is mediated through both CCK-A and CCK-B cholecystokinin receptors located most likely on the cell bodies of cholinergic interneurons in striatum.


Subject(s)
Acetylcholine/metabolism , Corpus Striatum/metabolism , Dopaminergic Neurons/physiology , Receptor, Cholecystokinin A/physiology , Receptor, Cholecystokinin B/physiology , Sincalide/pharmacology , Animals , Corpus Striatum/drug effects , Dopamine/pharmacology , Dopamine/physiology , Dopaminergic Neurons/drug effects , Male , Organ Culture Techniques , Rats , Rats, Wistar , Receptor, Cholecystokinin A/agonists , Receptor, Cholecystokinin B/agonists , Sincalide/physiology
9.
Bioorg Med Chem Lett ; 22(8): 2943-7, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22424974

ABSTRACT

New cholecystokinin-1 receptor (CCK1R) agonist 'triggers' were identified using iterative library synthesis. Structural activity relationship studies led to the discovery of compound 10e, a potent CCK1R agonist that demonstrated robust weight loss in a diet-induced obese rat model with very low systemic exposure. Pharmacokinetic data suggest that efficacy is primarily driven through activation of CCK1R's located within the intestinal wall.


Subject(s)
Amides/chemical synthesis , Drug Discovery , Piperidines/chemical synthesis , Receptor, Cholecystokinin A/agonists , Amides/chemistry , Amides/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , Humans , Inhibitory Concentration 50 , Male , Mice , Mice, Obese , Piperidines/chemistry , Piperidines/pharmacology , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Weight Loss/drug effects
10.
Bioorg Med Chem Lett ; 21(10): 2911-5, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21493064

ABSTRACT

A series of six-membered heterocycle carboxamides were synthesized and evaluated as cholecystokinin 1 receptor (CCK1R) agonists. A pyrimidine core proved to be the best heterocycle, and SAR studies resulted in the discovery of analog 5, a potent and structurally diverse CCK1R agonist.


Subject(s)
Amides/chemical synthesis , Amides/pharmacology , Receptor, Cholecystokinin A/agonists , Amides/chemistry , Animals , Cells, Cultured , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Humans , Inhibitory Concentration 50 , Mice , Molecular Structure , Protein Binding/drug effects , Pyrimidines/chemistry , Structure-Activity Relationship
11.
Am J Physiol Gastrointest Liver Physiol ; 300(2): G217-27, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21109591

ABSTRACT

The paradigm for the control of feeding behavior has changed significantly. Research has shown that leptin, in the presence of CCK, may mediate the control of short-term food intake. This interaction between CCK and leptin occurs at the vagus nerve. In the present study, we aimed to characterize the interaction between CCK and leptin in the vagal primary afferent neurons. Single neuronal discharges of vagal primary afferent neurons innervating the gastrointestinal tract were recorded from rat nodose ganglia. Three groups of nodose ganglia neurons were identified: group 1 responded to CCK-8 but not leptin; group 2 responded to leptin but not CCK-8; group 3 responded to high-dose CCK-8 and leptin. In fact, the neurons in group 3 showed CCK-8 and leptin potentiation, and they responded to gastric distention. To identify the CCK-A receptor (CCKAR) affinity states that colocalize with the leptin receptor OB-Rb, we used CCK-JMV-180, a high-affinity CCKAR agonist and low-affinity CCKAR antagonist. As expected, immunohistochemical studies showed that CCK-8 administration significantly potentiated the increase in the number of c-Fos-positive neurons stimulated by leptin in vagal nodose ganglia. Administration of CCK-JMV-180 eliminated the synergistic interaction between CCK-8 and leptin. We conclude that both low- and high-affinity CCKAR are expressed in nodose ganglia. Many nodose neurons bearing low-affinity CCKAR express OB-Rb. These neurons also respond to mechanical distention. An interaction between CCKAR and OB-Rb in these neurons likely facilitates leptin mediation of short-term satiety.


Subject(s)
Leptin/metabolism , Nodose Ganglion/metabolism , Receptor, Cholecystokinin A/metabolism , Receptors, Leptin/metabolism , Satiety Response/physiology , Animals , Binding, Competitive , Compliance/physiology , Drug Synergism , Electrophysiological Phenomena , Leptin/pharmacology , Male , Neurons, Afferent/classification , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Nodose Ganglion/cytology , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A/agonists , Receptor, Cholecystokinin A/antagonists & inhibitors , Sincalide/analogs & derivatives , Sincalide/pharmacology , Stomach/physiology , Time Factors , Tissue Distribution , Vagus Nerve/cytology , Vagus Nerve/metabolism
12.
Pancreas ; 39(2): 127-34, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19959964

ABSTRACT

OBJECTIVES: This study evaluated the role played by cholecystokinin (CCK) receptors' occupation in the control of somatostatin (SS) secretion in RIN-14B cells. METHODS: The presence of the CCK receptors 1 and 2 was confirmed by immunofluorescence, and SS secretion was evaluated by enzyme-linked immunosorbent assay. RESULTS: By immunofluorescence, 95% of the cell population was composed of SS cells bearing both CCK-R subtypes with 5% of beta cells (data not shown). Cerulein (Cae), a CCK-1R agonist, and pentagastrin, a CCK-2R agonist, dose-dependently increased SS release, 3-fold at 1 mumol/L Cae, 2.5-fold at 10 mumol/L pentagastrin, with occupation of both CCKRs confirmed by L-364,178 and L-365,260 inhibition of CCK receptors 1 and 2. The occupation of high-affinity CCK-1R by Cae was confirmed on SS release with JMV-180, a high-affinity CCK-1R agonist, and absence of SS release inhibition at high Cae concentration occupying the low-affinity CCK-1R. These cells release more than 60% of their SS content by constitutive secretion, confirmed by cycloheximide and brefeldin inhibiting SS synthesis and intracellular trafficking, respectively. CONCLUSIONS: Both CCKR subtypes occupy RIN-14B cells and initiate SS secretion through constitutive secretion controlled at SS synthesis level. Somatostatin secretion via the CCK-1R occupation mobilizes its high-affinity sites.


Subject(s)
Islets of Langerhans/metabolism , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin B/metabolism , Somatostatin/metabolism , Animals , Benzodiazepinones/pharmacology , Brefeldin A/pharmacology , Cell Line , Ceruletide/pharmacology , Cholecystokinin/metabolism , Cycloheximide/pharmacology , Devazepide/pharmacology , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gastrins/metabolism , Islets of Langerhans/drug effects , Pentagastrin/pharmacology , Phenylurea Compounds/pharmacology , Protein Precursors/metabolism , Protein Synthesis Inhibitors/pharmacology , Protein Transport , Rats , Receptor, Cholecystokinin A/agonists , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin B/agonists , Receptor, Cholecystokinin B/antagonists & inhibitors , Sincalide/analogs & derivatives , Sincalide/pharmacology , Somatostatin/biosynthesis
13.
Clin Pharmacol Ther ; 85(4): 362-5, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19295535

ABSTRACT

There are two interacting components in a clinical trial: the drug and the study design. When a trial does not work, we blame the drug--and the study is usually not published. This Commentary provides a context for the use of efficacy pipeline pharmacogenetics (PGx) in therapeutic programs. Jordan et al. published the results of an obesity trial with a cholecystokinin-A (CCK-A) receptor agonist and concluded that CCK-A by itself does not have a central role in long-term energy balance. The conclusions were sound, the report accurate, and the journal commendable for publishing a negative study, but the trial design was misdirected--it did not build on phase IIA information and did not test the proposed mechanism of action. The hypotheses should have been based on the original putative role of a central mechanism affecting appetite, which had been validated using efficacy PGx in phase IIA.


Subject(s)
Benzodiazepines/pharmacology , Eating/genetics , Pharmacogenetics/methods , Randomized Controlled Trials as Topic/methods , Receptor, Cholecystokinin A/genetics , Weight Loss/genetics , Eating/drug effects , Humans , Receptor, Cholecystokinin A/agonists
14.
Bioorg Med Chem Lett ; 18(17): 4833-7, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18684621

ABSTRACT

The discovery and structure-activity relationship of 1,2-diarylimidazole piperazine carboxamides bearing polar side chains as potent and selective cholecystokinin 1 receptor (CCK1R) agonists are described. Optimization of this series resulted in the discovery of isopropyl carboxamide 40, a CCK1R agonist with sub-nanomolar functional and binding activity as well as excellent potency in a mouse overnight food intake reduction assay.


Subject(s)
Anti-Obesity Agents/pharmacology , Benzodiazepines/pharmacology , Indoles/pharmacology , Obesity/drug therapy , Receptor, Cholecystokinin A/agonists , Thiazoles/pharmacology , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/chemistry , Benzodiazepines/chemical synthesis , Benzodiazepines/chemistry , Chemokines, CC , Humans , Indoles/chemical synthesis , Indoles/chemistry , Methylamines/chemical synthesis , Methylamines/chemistry , Methylamines/pharmacology , Mice , Piperazine , Piperazines/chemistry , Receptors, Cholecystokinin/agonists , Receptors, Cholecystokinin/chemistry , Thiazoles/chemical synthesis , Thiazoles/chemistry
15.
J Mol Model ; 14(4): 303-14, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18286312

ABSTRACT

Pyridopyrimidine-based analogues are among the most highly potent and selective antagonists of cholecystokinin receptor subtype-1 (CCK1R) described to date. To better understand the structural and chemical features responsible for the recognition mechanism, and to explore the binding pocket of these compounds, we performed automated molecular docking using GOLD2.2 software on some derivatives with structural diversity, and propose a putative binding conformation for each compound. The docking protocol was guided by the key role of the Asn333 residue, as revealed by site directed mutagenesis studies. The results suggest two putative binding modes located in the same pocket. Both are characterized by interaction with the main residues revealed by experiment, Asn333 and Arg336, and differ in the spatial position of the Boc-Trp moiety of these compounds. Hydrophobic contacts with residues Thr117, Phe107, Ile352 and Ile329 are also in agreement with experimental data. Despite the poor correlation obtained between the estimated binding energies and the experimental activity, the proposed models allow us to suggest a plausible explanation of the observed binding data in accordance with chemical characteristics of the compounds, and also to explain the observed diastereoselectivity of this family of antagonists towards CCK1R. The most reasonable selected binding conformations could be the starting point for future studies. Figure Superimposition of the two putative binding conformations revealed by molecular docking for pyridopyrimidine-based CCK1 antagonists.


Subject(s)
Drug Design , Models, Molecular , Pyrimidines/chemistry , Receptor, Cholecystokinin A/chemistry , Binding Sites , Computer Simulation , Entropy , Ligands , Molecular Conformation , Protein Conformation , Pyrimidines/pharmacology , Receptor, Cholecystokinin A/agonists , Software , Stereoisomerism , Structure-Activity Relationship
16.
J Agric Food Chem ; 56(3): 837-43, 2008 Feb 13.
Article in English | MEDLINE | ID: mdl-18211011

ABSTRACT

Protein has been reported to be the most satiating of all macronutrients. Upon gastrointestinal digestion, peptides are generated that stimulate the release of satiety hormones such as cholecystokinin (CCK) from enteroendocrine cells. As such, bioactive peptides could be the target of Functional Food ingredients with satiating effects. We set up an in vitro assay system to investigate if different protein hydrolysates exhibit varying CCK-releasing properties. Soy, pea, potato, casein, and whey protein hydrolysates were incubated with the enteric endocrine cell line STC-1 that endogenously expresses and secretes CCK. Release of CCK was measured by ELISA. All hydrolysates induced CCK release at low concentrations (>0.1 mg.L -1)); however, no significant differences in CCK-releasing properties between the different protein hydrolysates were found, suggesting a generic, nonspecific peptide-sensing mechanism in the STC-1 cells on hydrolyzed protein. As the ELISA exhibits sensitivity to all CCK isoforms possessing the C-terminal CCK octapeptide but varying in biological activity at the CCK 1 receptor (CCK 1R), a secondary module was added to the STC-1 cell assay. Intracellular calcium measurements were performed in CHO-CCK 1R cells. Following exposure of the STC-1 cells to the protein hydrolysates, the medium was tested on the CCK 1R assay. Released CCK was measured with higher sensitivity and lower variability than in the ELISA. Surprisingly, we found that some protein hydrolysates (soy > potato >> casein) also directly stimulated CCK 1R-expressing cells, while whey and pea protein hydrolysates were inactive. As CCK 1R is expressed in the GI tract, direct interaction of CCK 1R with dietary peptides may contribute to their satiety effects. Future experiments developing bioactive ingredients for Functional Foods for weight management could involve isolation of the active, CCK 1R-activating peptides in, for example, soy protein hydrolysates.


Subject(s)
Cholecystokinin/metabolism , Enteroendocrine Cells/drug effects , Enteroendocrine Cells/metabolism , Protein Hydrolysates/pharmacology , Receptor, Cholecystokinin A/agonists , Animals , Arabidopsis Proteins , CHO Cells , Cell Line , Cricetinae , Cricetulus , Enzyme-Linked Immunosorbent Assay , Mice , Rats , Receptor, Cholecystokinin A/drug effects , Receptor, Cholecystokinin A/physiology , Recombinant Proteins/drug effects , Starch Synthase
17.
Clin Pharmacol Ther ; 83(2): 281-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17597711

ABSTRACT

Cholecystokinin (CCK) decreases meal size through activation of CCK-A receptors on vagal afferents. We tested the hypothesis that the selective CCK-A agonist GI181771X induces weight loss in obese patients. Patients with body mass index > or = 30 or > or = 27 kg/m2 with concomitant risk factors were randomized to 24-week, double-blind treatment with different GI181771X doses or matching placebo together with a hypocaloric diet. The primary efficacy end point was the absolute change in body weight. To monitor pancreatic and gallbladder effects, patients underwent abdominal ultrasound and magnetic resonance imaging before and after treatment. We randomized 701 patients to double-blind treatment. GI181771X did not reduce body weight and had no effect on waist circumference or other cardiometabolic risk markers. Gastrointestinal side effects were more common with GI181771X than with placebo treatment, whereas hepatobiliary or pancreatic abnormalities did not occur. CCK-A by itself does not have a central role in long-term energy balance.


Subject(s)
Anti-Obesity Agents/therapeutic use , Benzodiazepines/therapeutic use , Caloric Restriction , Obesity/drug therapy , Receptor, Cholecystokinin A/agonists , Weight Loss/drug effects , Adult , Anti-Obesity Agents/adverse effects , Benzodiazepines/adverse effects , Combined Modality Therapy , Double-Blind Method , Female , Gallbladder/drug effects , Humans , Liver/drug effects , Magnetic Resonance Imaging , Male , Middle Aged , Obesity/diet therapy , Obesity/metabolism , Obesity/pathology , Pancreas/drug effects , Receptor, Cholecystokinin A/metabolism , Time Factors , Treatment Outcome , Ultrasonography
18.
J Med Chem ; 49(3): 850-63, 2006 Feb 09.
Article in English | MEDLINE | ID: mdl-16451051

ABSTRACT

An understanding of the molecular basis of drug action provides opportunities for refinement of drug properties and for development of more potent and selective molecules that act at the same biological target. In this work, we have identified the active enantiomers in racemic mixtures of structurally related benzophenone derivatives of 1,5-benzodiazepines, representing both antagonist and agonist ligands of the type A cholecystokinin receptor. The parent compounds of the 1,5-benzodiazepine CCK receptor photoaffinity ligands were originally prepared in an effort to develop orally active drugs. The enantiomeric compounds reported in this study selectively photoaffinity-labeled the CCK receptor, resulting in the identification of a site of attachment for the photolabile moiety of the antagonist probe deep within the receptor's membrane-spanning region at Leu(88), a residue within transmembrane segment two. In contrast, the agonist probe labeled a region including extracellular loop one and a portion of transmembrane segment three. The antagonist covalent attachment site to the receptor served as a guide in the construction of theoretical three-dimensional molecular models for the antagonist-receptor complex. These models provided a means for visualization of physically plausible ligand-receptor interactions in the context of all currently available biological data that address small molecule interactions with the CCK receptor. Our approach, featuring the use of novel photolabile compounds targeting the membrane-spanning receptor domain to probe the binding site region, introduces powerful tools and a strategy for direct and selective investigation of nonpeptidyl ligand binding to peptide receptors.


Subject(s)
Benzodiazepines/chemical synthesis , Benzophenones/chemical synthesis , Photoaffinity Labels/chemical synthesis , Receptor, Cholecystokinin A/agonists , Receptor, Cholecystokinin A/antagonists & inhibitors , Animals , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Benzophenones/chemistry , Benzophenones/pharmacology , Binding Sites , CHO Cells , Cell Membrane/metabolism , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , Devazepide/chemistry , Devazepide/pharmacology , In Vitro Techniques , Ligands , Models, Molecular , Pancreas/cytology , Pancreas/drug effects , Pancreas/metabolism , Photoaffinity Labels/chemistry , Photoaffinity Labels/pharmacology , Protein Structure, Tertiary , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A/metabolism , Stereoisomerism , Structure-Activity Relationship
19.
Pancreatology ; 6(1-2): 65-75; discussion 75-6, 2006.
Article in English | MEDLINE | ID: mdl-16327284

ABSTRACT

BACKGROUND: We have investigated the involvement of cholecystokinin (CCK) receptor subtypes in haemodynamic changes in the pancreas of anaesthetized rats during submaximal and supramaximal stimulation with the CCK analogue, caerulein. METHODS: For submaximal stimulation, caerulein (0.4 nmol/kg/h) was infused intravenously, while acute pancreatitis was induced by intraperitoneal injections of high doses of caerulein (3 x 25 nmol/kg). Pancreatic blood flow was measured by hydrogen clearance. RESULTS: Low caerulein doses increased pancreatic blood flow by 26 +/- 8% and vascular conductance by 24 +/- 4%. This effect was mimicked by the CCK2 agonist gastrin-17. All effects were abolished by a CCK2 antagonist while a CCK1 antagonist remained inactive. Conversely, amylase output by caerulein was abolished by CCK1 receptor blockade, but not by inhibition of CCK2 receptors. During caerulein-induced pancreatitis, vascular conductance increased by 109 +/- 26% and remained elevated throughout the experiment; vascular flow initially increased by 62 +/- 27% and then returned to baseline. The vascular effects were prevented by a CCK2 receptor antagonist, while the induction of pancreatitis was due to CCK1 receptor stimulation. CONCLUSIONS: Caerulein increases pancreatic vascular flow via activation of CCK2 receptors. This effect occurs both at submaximal and at supramaximal levels of exocrine stimulation.


Subject(s)
Ceruletide/pharmacology , Pancreas, Exocrine/drug effects , Receptors, Cholecystokinin/metabolism , Animals , Bradykinin B2 Receptor Antagonists , Female , Gastrins/pharmacology , Indoles/pharmacology , Pancreas, Exocrine/blood supply , Pancreatitis/chemically induced , Pancreatitis/pathology , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A/agonists , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin B/agonists , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptors, Cholecystokinin/drug effects , Regional Blood Flow/drug effects , Stimulation, Chemical
20.
J Pharmacol Toxicol Methods ; 54(1): 36-41, 2006.
Article in English | MEDLINE | ID: mdl-16246596

ABSTRACT

INTRODUCTION: Cholecystokinin type-1 (CCK(1)) receptors mediate many of the physiological functions of CCK including delay of gastric emptying, pancreatic enzyme secretion, intestinal motility and gallbladder contractility. Existing in-vivo assays for the quantitative measurement of CCK(1) receptor mediated function are generally variable, limited in precision and require a relatively large number of animals to obtain statistically meaningful data. We found that they did not provide robust pharmacokinetic-pharmacodynamic data for profiling compounds acting at these receptors. Accordingly, here we describe a novel rat duodenal contractility assay that addresses these problems. METHODS: Rats were anaesthetised and a saline-filled balloon was inserted through the body of the stomach and secured in the duodenum approximately 1 cm from the pyloric sphincter for measurement of intra-lumenal pressure. Studies were performed to determine a dose, rate and frequency of administration of CCK8S that produced a readily quantifiable response. RESULTS: Initial experiments revealed that sustained exposure to CCK8S resulted in the rapid development of tachyphylaxis. After investigating different dosing paradigms, it was found that pulsatile delivery of CCK8S (intravenous infusion for 1 min every 10 min) produced a readily quantifiable contractile response that did not exhibit tachyphylaxis. The assay response output was defined as the number of contractions >5 mm Hg over baseline. The contractions were blocked in a dose-dependent manner by intravenous bolus injections of the CCK(1) receptor antagonists, dexloxiglumide (2 and 20 micromol/kg), and devazepide (3-100 nmol/kg) but not by the CCK(2) receptor antagonist gastrazole (10 micromol/kg). CONCLUSION: A novel, simple, high quality assay for the quantification of the in-vivo activity of CCK(1) receptor ligands is described. CCK8S delivered by pulsatile intravenous infusion to anesthetized rats produced a burst of contractile activity of the duodenum mediated by CCK(1) receptors. This activity was highly reproducible and sustained for more than 3 h providing an assay that circumvents problems associated with agonist-induced tachyphylaxis.


Subject(s)
Anesthesia/methods , Biological Assay/methods , Receptor, Cholecystokinin A/metabolism , Animals , Dose-Response Relationship, Drug , Duodenum/drug effects , Duodenum/physiology , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A/agonists , Receptor, Cholecystokinin A/antagonists & inhibitors , Sincalide/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...