Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 748
Filter
1.
Nat Chem Biol ; 17(12): 1230-1237, 2021 12.
Article in English | MEDLINE | ID: mdl-34556863

ABSTRACT

Cholecystokinin receptors, CCKAR and CCKBR, are important neurointestinal peptide hormone receptors and play a vital role in food intake and appetite regulation. Here, we report three crystal structures of the human CCKAR in complex with different ligands, including one peptide agonist and two small-molecule antagonists, as well as two cryo-electron microscopy structures of CCKBR-gastrin in complex with Gi2 and Gq, respectively. These structures reveal the recognition pattern of different ligand types and the molecular basis of peptide selectivity in the cholecystokinin receptor family. By comparing receptor structures in different conformational states, a stepwise activation process of cholecystokinin receptors is proposed. Combined with pharmacological data, our results provide atomic details for differential ligand recognition and receptor activation mechanisms. These insights will facilitate the discovery of potential therapeutics targeting cholecystokinin receptors.


Subject(s)
Devazepide/chemistry , Receptors, Cholecystokinin/chemistry , Amino Acid Sequence , Cryoelectron Microscopy , Crystallization , Humans , Indoleacetic Acids/chemistry , Ligands , Models, Molecular , Protein Binding , Protein Conformation , Receptors, Cholecystokinin/genetics , Structure-Activity Relationship , Thiazoles/chemistry
2.
Am J Physiol Endocrinol Metab ; 315(1): E81-E90, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29533738

ABSTRACT

The idea that gut-derived satiation signals influence food reward has recently gained traction, but this hypothesis is largely based on studies focused on neural circuitry, not the peripherally released signals. Here, we directly tested the hypothesis that intragastric (IG) nutrient infusion can suppress motivation for food. In a series of experiments, IG sucrose infusion (15 kcal) significantly and reliably reduced operant responding for a sucrose reward on a progressive ratio (PR) schedule. Moreover, food deprivation for 24 h before the test session did not prevent the suppressive effect of nutrients. The suppressive effect of IG sucrose on fixed ratio 5 (FR5) operant responding was also assessed as a comparison. The effect of IG nutrients to reduce motivation was not limited to sucrose; IG Ensure infusion (9.3 kcal) also significantly reduced PR operant responding for sucrose pellets. To verify that these effects were not secondary to the osmotic challenge of concentrated nutrients, we tested IG infusion of noncaloric saline solutions equiosmolar to 40% sucrose or Ensure and found no effect. Finally, we focused on glucagon-like peptide-1 (GLP-1) and cholecystokinin (CCK) as candidate mediators for the effect of IG nutrients. Pretreatment with exendin-9, a GLP-1 receptor antagonist, delivered intraperitoneally, significantly attenuated the ability of IG nutrients to suppress PR responding and breakpoint in males, but not in females, whereas pretreatment with devazepide, a CCKA receptor antagonist, failed to do so in both sexes. Together, these data support the idea that nutrient-induced satiation signals influence food reward and may implicate GLP-1 in this process.


Subject(s)
Enteral Nutrition/psychology , Motivation , Animals , Cholecystokinin/metabolism , Conditioning, Operant , Devazepide/pharmacology , Estrous Cycle/drug effects , Female , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Intubation, Gastrointestinal , Male , Rats , Rats, Wistar , Receptor, Cholecystokinin A/antagonists & inhibitors , Reinforcement Schedule , Reward , Sucrose/pharmacology
3.
Eur J Clin Invest ; 46(2): 158-69, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26683129

ABSTRACT

BACKGROUND: A defect in gallbladder contraction function plays a key role in the pathogenesis of gallstones. The cholecystokinin-1 receptor (CCK-1R) antagonists have been extensively investigated for their therapeutic effects on gastrointestinal and metabolic diseases in animal studies and clinical trials. However, it is still unknown whether they have a potential effect on gallstone formation. DESIGN: To study whether the CCK-1R antagonists enhance cholelithogenesis, we investigated cholesterol crystallization, gallstone formation, hepatic lipid secretion, gallbladder emptying function and intestinal cholesterol absorption in male C57BL/6J mice treated by gavage with devazepide (4 mg/day/kg) or vehicle (as controls) twice per day and fed the lithogenic diet for 21 days. RESULTS: During 21 days of feeding, oral administration of devazepide significantly accelerated cholesterol crystallization and crystal growth to microlithiasis, with 40% of mice forming gallstones, whereas only agglomerated cholesterol monohydrate crystals were found in mice receiving vehicle. Compared to the vehicle group, fasting and postprandial residual gallbladder volumes in response to the high-fat meal were significantly larger in the devazepide group during cholelithogenesis, showing reduced gallbladder emptying and bile stasis. Moreover, devazepide significantly increased hepatic secretion of biliary cholesterol, but not phospholipids or bile salts. The percentage of intestinal cholesterol absorption was higher in devazepide-treated mice, increasing the bioavailability of chylomicron-derived cholesterol in the liver for biliary hypersecretion into bile. These abnormalities induced supersaturated bile and rapid cholesterol crystallization. CONCLUSIONS: The potent CCK-1R antagonist devazepide increases susceptibility to gallstone formation by impairing gallbladder emptying function, disrupting biliary cholesterol metabolism and enhancing intestinal cholesterol absorption in mice.


Subject(s)
Cholelithiasis/chemically induced , Cholesterol/metabolism , Devazepide/pharmacology , Gallbladder Emptying/drug effects , Gallbladder/drug effects , Hormone Antagonists/pharmacology , Intestines/drug effects , Receptor, Cholecystokinin A/antagonists & inhibitors , Animals , Bile Acids and Salts/metabolism , Cholelithiasis/metabolism , Gallbladder/metabolism , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Receptor, Cholecystokinin A/drug effects , Receptor, Cholecystokinin A/genetics
4.
Am J Physiol Gastrointest Liver Physiol ; 306(3): G173-82, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24264048

ABSTRACT

Gastric emptying and gastric secretion are two major physiological functions of the stomach. The assessment of these functions in particular in small animals is challenging; no method currently available allows the simultaneous measurement of both functions, and methods used are lethal or invasive and often limited by spatial, temporal, or quantitative resolution. Here, we report the establishment and validation of a quantitative noninvasive high-throughput computed tomography-based method to measure simultaneously gastric emptying and secretion in rats in vivo. The imaging strategy enables one to visualize stomach anatomy and to quantify stomach volume and stomach contrast agent content. The method was validated by comparing the results to classical lethal methods (stomach phenol red content and stomach wet weight). Additionally, we showed that the use of a mild anesthetic does not interfere with normal gastric function, thereby enabling high-resolution temporal studies within single animals. These combined advantages were applied to reevaluate the impact of cholecystokinin (CCK), histamine, and oral glucose solutions on gastric function with high temporal resolution. CCK inhibited gastric emptying completely for 20 min, leading to the accumulation of gastric juice in the stomach. The CCK antagonist devazepide blocked this effect. Histamine stimulated both gastric secretion and delayed emptying. Oral glucose solution emptied at a fixed rate of 24-31 cal/min and stimulated gastric secretion. These results confirm previous observations and add volumetric changes as a new dimension. As computed tomography scanners become broadly available, this method is an excellent approach to measure the combined gastric functional readout and to reduce the number of animals used.


Subject(s)
Cholecystokinin/pharmacology , Devazepide/pharmacology , Gastric Emptying/drug effects , Stomach/drug effects , Tomography, X-Ray Computed/methods , Animals , Cholecystokinin/antagonists & inhibitors , Gastric Emptying/physiology , Histamine/pharmacology , Male , Models, Animal , Rats , Rats, Wistar , Stomach/physiology
5.
Am J Physiol Regul Integr Comp Physiol ; 306(5): R363-73, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24430886

ABSTRACT

The subfornical organ (SFO) is an important sensory circumventricular organ implicated in the regulation of fluid homeostasis and energy balance. We investigated whether the SFO is activated by the hormone cholecystokinin (CCK). CCK1 and CCK2 receptors were identified in the SFO by RT-PCR. Dissociated SFO neurons that responded to CCK (40/77), were mostly depolarized (9.2 ± 0.9 mV, 30/77), but some were hyperpolarized (-7.3 ± 1.1 mV, 10/77). We next examined the responses of SFO neurons in vivo to CCK (16 µg/kg ip), in the presence and absence of CCK1 or CCK2 receptor antagonists (devazepide; 600 µg/kg and L-365,260; 100 µg/kg, respectively), using the functional activation markers c-Fos and phosphorylated extracellular signal-related kinase (p-ERK). The nucleus of the solitary tract (NTS) served as a control for CCK-induced activity. There was a significant increase in c-Fos expression in the NTS (259.2 ± 20.8 neurons) compared with vehicle (47.5 ± 2.5). Similarly, in the SFO, c-Fos was expressed in 40.5 ± 10.6 neurons in CCK-treated compared with 6.6 ± 2.7 in vehicle-treated rats (P < 0.01). Devazepide significantly reduced the effects of CCK in the NTS but not in SFO. L-365,260 blocked the effects of CCK in both brain regions. CCK increased the number of p-ERK neurons in NTS (27.0 ± 4.0) as well as SFO (18.0 ± 4.0), compared with vehicle (8.0 ± 2.6 and 4.3 ± 0.6, respectively; P < 0.05). Both devazepide and L-365,260 reduced CCK-induced p-ERK in NTS, but only L-365,260 reduced it in the SFO. In conclusion, the SFO represents a novel brain region at which circulating CCK may act via CCK2 receptors to influence central autonomic control.


Subject(s)
Cholecystokinin/pharmacology , Peptide Fragments/pharmacology , Subfornical Organ/drug effects , Animals , Benzodiazepinones/pharmacology , Devazepide/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/physiology , Genes, fos/genetics , Genes, fos/physiology , Hormone Antagonists/pharmacology , Male , Neurons/drug effects , Neurons/metabolism , Phenylurea Compounds/pharmacology , RNA/genetics , RNA/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/genetics , Receptors, Cholecystokinin/metabolism , Subfornical Organ/cytology , Subfornical Organ/physiology
6.
Dig Dis Sci ; 59(6): 1180-91, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24817409

ABSTRACT

BACKGROUND: Obesity and dietary fat are associated with increased risk of several malignancies including pancreatic cancer. The incidence of pancreatic cancer is increased in countries that consume diets high in fat. AIM: The purpose of this study was to assess the relationship and mechanism of action between dietary fat and endogenous cholecystokinin (CCK) on pancreatic tumor growth and metastasis in an immunocompetent animal model. METHODS: C57BL/6 mice were placed on regular, low-fat, or high-fat diets for 8 weeks before establishment of Panc-02 orthotopic pancreatic tumors. Mice were then treated with a CCK-A receptor antagonist, devazepide, or vehicle for an additional 2.5 weeks. Pancreas tumors were weighed and metastases counted. Blood CCK levels were measured by radioimmunoassay (RIA). Tissues were examined histologically and studied for genes associated with metastasis by RT-PCR array. Effects of the CCK antagonist on Panc-02 cells invasiveness was assessed in a Matrigel invasion assay. RESULTS: Mice that received the high-fat diet had larger tumors and tenfold higher serum CCK levels by RIA compared to normal diet controls (p < 0.01). Pancreatic tumors in high-fat diet mice treated with the antagonist had fewer intravascular tumor emboli and metastases compared to controls. The reduction in tumor emboli correlated with decreased vascular endothelial growth factor-A (VEGF-A) expression in tumors (p < 6 × 10(-9)). In vitro invasiveness of Panc-02 cells also was reduced by CCK-A receptor antagonist treatment (p = 1.33 × 10(-6)). CONCLUSION: CCK is a mediator of dietary fat-associated pancreatic cancer. CCK is also involved in the invasiveness of pancreatic tumors through a mechanism involving VEGF-A.


Subject(s)
Cholecystokinin/metabolism , Dietary Fats/adverse effects , Pancreatic Neoplasms/metabolism , Animals , Blood Glucose , Cell Line, Tumor , Devazepide/pharmacology , Dietary Fats/administration & dosage , Dose-Response Relationship, Drug , Embolism/prevention & control , Hormone Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Neoplasm Metastasis/prevention & control , Pancreatic Neoplasms/pathology , Radioimmunoassay
7.
Neurosci Lett ; 818: 137557, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37972685

ABSTRACT

The study was performed to evaluate the role of central serotoninergic, GABAergic, and cholecystokinin systems in neuropeptide VF (NPVF)-induced hypophagia in broiler chickens. In this study, 9 experiments were designed, each with one control and three treatment groups (n = 44 in each experiment). Control chicks of all groups were subjected to normal saline + Evans blue 0.1 % Intracerebroventricular (ICV) injection. In the first experiment, 3 groups of chicks received NPVF (4, 8, and 16 nmol). In experiment 2-9, one group of chicks received NPVF (16 nmol), another received 10 µg fluoxetine (serotonin reuptake inhibitor) (experiment 2), 1.25 µg PCPA (serotonin synthesis inhibitor) (experiment 3), 1.5 µg SB-242,084 (5-HT2C receptor antagonist) (experiment 4), 15.25 nmol 8-OH-DPAT (5-HT1A receptor antagonist) (experiment 5), 0.5 µg picrotoxin (GABAA receptor antagonist) (experiment 6), 20 ng CGP54626 (GABAB receptor antagonist) (experiment 7), 1 nmol devazepide (CCKA receptor antagonist) (experiment 8), and 1 nmol/L-365(-|-),260 (CCKB receptor antagonist) (experiment 9), and another final group received combination of specific neurotransmitter + NPVF Then, the cumulative food intake was measured until 120 min post-injection. ICV injection of NPVF (8 and 16 nmol) significantly decreased food intake (P < 0.05). Simultaneous injection of fluoxetine + NPVF and also picrotoxin + NPVF significantly increased hypophagia caused by NPVF (P < 0.05). However, co-administration of PCPA + NPVF and also SB242084 + NPVF significantly decreased NPVF-induced hypophagia (P < 0.05). Finally, 8-OH-DPAT, CGP54626, devazepide, and L-365,260 had no effect on the hypophagia brought on by NPVF (P > 0.05). Count-type behaviors were dose-dependent and decreased in groups that received NPVF compared to the control group (P < 0.05). Our finding recommended an interconnection between central NPVF and serotoninergic, GABAergic, and cholecystokinin systems in neonatal chickens.


Subject(s)
Chickens , Cholecystokinin , Feeding Behavior , Animals , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Cholecystokinin/pharmacology , Devazepide/pharmacology , Eating , Fluoxetine/pharmacology , Picrotoxin/pharmacology , Serotonin Antagonists/pharmacology
8.
J Neurosci ; 32(48): 17097-107, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23197703

ABSTRACT

Leptin is an adipocyte-derived hormone that controls energy balance by acting primarily in the CNS, but its action is lost in common forms of obesity due to central leptin resistance. One potential mechanism for such leptin resistance is an increased hypothalamic expression of Suppressor of cytokine signaling 3 (Socs3), a feedback inhibitor of the Jak-Stat pathway that prevents Stat3 activation. Ample studies have confirmed the important role of Socs3 in leptin resistance and obesity. However, the degree to which Socs3 participates in the regulation of energy homeostasis in nonobese conditions remains largely undetermined. In this study, using adult mice maintained under standard diet, we demonstrate that Socs3 deficiency in the mediobasal hypothalamus (MBH) reduces food intake, protects against body weight gain, and limits adiposity, suggesting that Socs3 is necessary for normal body weight maintenance. Mechanistically, MBH Socs3-deficient mice display increased hindbrain sensitivity to endogenous, meal-related satiety signals, mediated by oxytocin signaling. Thus, oxytocin signaling likely mediates the effect of hypothalamic leptin on satiety circuits of the caudal brainstem. This provides an anatomical substrate for the effect of leptin on meal size, and more generally, a mechanism for how the brain controls short-term food intake as a function of the energetic stores available in the organism to maintain energy homeostasis. Any dysfunction in this pathway could potentially lead to overeating and obesity.


Subject(s)
Hypothalamus/metabolism , Oxytocin/metabolism , Rhombencephalon/metabolism , Satiety Response/physiology , Signal Transduction/physiology , Suppressor of Cytokine Signaling Proteins/genetics , Animals , Devazepide/pharmacology , Eating/drug effects , Eating/physiology , Hormone Antagonists/pharmacology , Hypothalamus/drug effects , Leptin/metabolism , Mice , Receptors, Cholecystokinin/antagonists & inhibitors , Rhombencephalon/drug effects , Satiety Response/drug effects , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism
9.
Am J Physiol Regul Integr Comp Physiol ; 305(4): R404-13, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23785073

ABSTRACT

Acupuncture or electroacupuncture (EA) potentially offers a nonpharmacological approach to reduce high blood pressure (BP). However, ~70% of the patients and animal subjects respond to EA, while 30% do not. EA acts, in part, through an opioid mechanism in the rostral ventrolateral medulla (rVLM) to inhibit sympathoexcitatory reflexes induced by gastric distention. CCK-8 opposes the action of opioids during analgesia. Therefore, we hypothesized that CCK-8 in the rVLM antagonizes EA modulation of sympathoexcitatory cardiovascular reflex responses. Male rats anesthetized with ketamine and α-chloralose subjected to repeated gastric distension every 10 min were examined for their responsiveness to EA (2 Hz, 0.5 ms, 1-4 mA) at P5-P6 acupoints overlying median nerve. Repeated gastric distension every 10 min evoked consistent sympathoexcitatory responses. EA at P5-P6 modulated gastric distension-induced responses. Microinjection of CCK-8 in the rVLM reversed the EA effect in seven responders. The CCK1 receptor antagonist devazepide microinjected into the rVLM converted six nonresponders to responders by lowering the reflex response from 21 ± 2.2 to 10 ± 2.9 mmHg (first vs. second application of EA). The EA modulatory action in rats converted to responders with devazepide was reversed with rVLM microinjection of naloxone (n = 6). Microinjection of devazepide in the absence of a second application of EA did not influence the primary pressor reflexes of nonresponders. These data suggest that CCK-8 antagonizes EA modulation of sympathoexcitatory cardiovascular responses through an opioid mechanism and that inhibition of CCK-8 can convert animals that initially are unresponsive to EA to become responsive.


Subject(s)
Blood Pressure , Electroacupuncture , Hypertension/prevention & control , Mechanotransduction, Cellular , Medulla Oblongata/metabolism , Reflex , Sincalide/metabolism , Stomach/innervation , Animals , Blood Pressure/drug effects , Devazepide/administration & dosage , Disease Models, Animal , Enkephalins/metabolism , Hormone Antagonists/administration & dosage , Hypertension/etiology , Hypertension/metabolism , Hypertension/physiopathology , Male , Medulla Oblongata/drug effects , Medulla Oblongata/physiopathology , Microinjections , Narcotic Antagonists/administration & dosage , Pressure , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin A/metabolism , Sincalide/administration & dosage , Time Factors
10.
Horm Behav ; 64(5): 812-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24100196

ABSTRACT

Deficits in satiation signaling during obesogenic feeding have been proposed to play a role in hyperphagia and weight gain in animals prone to become obese. However, whether this impaired signaling is due to high fat (HF) feeding or to their obese phenotype is still unknown. Therefore, in the current study, we examined the effects of CCK-8 (0.5, 1.0, 2.0, and 4.0 µg/kg) on suppression of food intake of HF-fed obese prone (OP) and resistant (OR) rats. Additionally, we determined the role of endogenous CCK in lipid-induced satiation by measuring plasma CCK levels following a lipid gavage, and tested the effect of pretreatment with devazepide, a CCK-1R antagonist on intragastric lipid-induced satiation. Finally, we examined CCK-1R mRNA levels in the nodose ganglia. We show that OP rats have reduced feeding responses to the low doses of exogenous CCK-8 compared to OR rats. Furthermore, OP rats exhibit deficits in endogenous CCK signaling, as pretreatment with devazepide failed to abolish the reduction in food intake following lipid gavage. These effects were associated with reduced plasma CCK after intragastric lipid in OP but not OR rats. Furthermore, HF feeding resulted in downregulation of CCK-1Rs in the nodose ganglia of OP rats. Collectively, these results demonstrate that HF feeding leads to impairments in lipid-induced CCK satiation signaling in obese-prone rats, potentially contributing to hyperphagia and weight gain.


Subject(s)
Cholecystokinin/metabolism , Diet, High-Fat , Obesity/metabolism , Animals , Cholecystokinin/antagonists & inhibitors , Devazepide/pharmacology , Dietary Fats/pharmacology , Down-Regulation/drug effects , Eating/drug effects , Feeding Behavior/drug effects , Hormone Antagonists/pharmacology , Male , Rats , Signal Transduction/drug effects , Sincalide/pharmacology
11.
Appetite ; 65: 1-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23376733

ABSTRACT

We have analyzed the effect of palmitoleic acid on short-term food intake in male rats. Administration of omega-7 palmitoleic acid by oral gavage significantly decreased food intake compared to palmitic acid, omega-9 oleic acid, or a vehicle control. Palmitoleic acid exhibited a dose-dependent effect in this context and did not cause general malaise. A triglyceride form of palmitoleate also decreased food intake, whereas olive oil, which is rich in oleic acid, did not. Palmitoleic acid accumulated within the small intestine in a dose-dependent fashion and elevated levels of the satiety hormone cholecystokinin (CCK). Both protein and mRNA levels of CCK were affected in this context. The suppression of food intake by palmitoleic acid was attenuated by intravenous injection of devazepide, a selective peripheral CCK receptor antagonist. Palmitoleic acid did not alter the expression of peroxisome proliferator-activated receptor alpha (PPARα) target genes, and a PPARα antagonist did not affect palmitoleic acid-induced satiety. This suggests that the PPARα pathway might not be involved in suppressing food intake in response to palmitoleic acid. We have shown that orally administered palmitoleic acid induced satiety, enhanced the release of satiety hormones in rats.


Subject(s)
Appetite/drug effects , Cholecystokinin/metabolism , Energy Intake/drug effects , Fatty Acids, Monounsaturated/pharmacology , Satiety Response/drug effects , Administration, Oral , Animals , Cholecystokinin/genetics , Devazepide/pharmacology , Dose-Response Relationship, Drug , Gene Expression/drug effects , Hormone Antagonists/pharmacology , Intestine, Small/drug effects , Intestine, Small/metabolism , Male , PPAR alpha/genetics , PPAR alpha/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Satiation/drug effects
12.
Am J Physiol Regul Integr Comp Physiol ; 303(12): R1231-40, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23115121

ABSTRACT

CCK is hypothesized to inhibit meal size by acting at CCK1 receptors (CCK1R) on vagal afferent neurons that innervate the gastrointestinal tract and project to the hindbrain. Earlier studies have shown that obese Otsuka Long-Evans Tokushima Fatty (OLETF) rats, which carry a spontaneous null mutation of the CCK1R, are hyperphagic and obese. Recent findings show that rats with CCK1R-null gene on a Fischer 344 background (Cck1r(-/-)) are lean and normophagic. In this study, the metabolic phenotype of this rat strain was further characterized. As expected, the CCK1R antagonist, devazepide, failed to stimulate food intake in the Cck1r(-/-) rats. Both Cck1r(+/+) and Cck1r(-/-) rats became diet-induced obese (DIO) when maintained on a high-fat diet relative to chow-fed controls. Cck1r(-/-) rats consumed larger meals than controls during the dark cycle and smaller meals during the light cycle. These effects were accompanied by increased food intake, total spontaneous activity, and energy expenditure during the dark cycle and an apparent reduction in respiratory quotient during the light cycle. To assess whether enhanced responsiveness to anorexigenic factors may contribute to the lean phenotype, we examined the effects of melanotan II (MTII) on food intake and body weight. We found an enhanced effect of MTII in Cck1r(-/-) rats to suppress food intake and body weight following both central and peripheral administration. These results suggest that the lean phenotype is potentially driven by increases in total spontaneous activity and energy expenditure.


Subject(s)
Energy Metabolism/physiology , Motor Activity/physiology , Phenotype , Receptor, Cholecystokinin A/deficiency , Thinness/physiopathology , Animals , Body Weight/drug effects , Body Weight/physiology , Devazepide/pharmacology , Eating/drug effects , Eating/physiology , Gene Deletion , Male , Models, Animal , Peptides, Cyclic/pharmacology , Rats , Rats, Inbred F344 , Rats, Mutant Strains , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin A/genetics , Sequence Deletion/genetics , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
13.
Biosci Biotechnol Biochem ; 76(6): 1104-9, 2012.
Article in English | MEDLINE | ID: mdl-22790930

ABSTRACT

We have recently reported that oral gavage of a potato extract (Potein®) suppressed the food intake in rats. The satiating effect of the potato extract was compared in the present study to other protein sources, and the involvement of endogenous cholecystokinin (CCK) secretion was examined. Food consumption was measured in 18-h fasted rats after oral gavage of the potato extract or other protein sources. The CCK-releasing activity of the potato extract was then examined in anesthetized rats with a portal cannula. Oral gavage of the potato extract reduced the food intake in the rats, the effect being greater than with casein and a soybean ß-conglycinin hydrolysate. The suppressive effect on appetite of the potato extract was attenuated by treating with a CCK-receptor antagonist (devazepide). The portal CCK concentration was increased after a duodenal administration of the potato extract to anesthetized rats. These results indicate that the potato extract suppressed the food intake in rats through CCK secretion.


Subject(s)
Appetite/drug effects , Cholecystokinin/metabolism , Eating/drug effects , Plant Extracts/administration & dosage , Solanum tuberosum/chemistry , Administration, Oral , Animals , Antigens, Plant/pharmacology , Appetite/physiology , Caseins/pharmacology , Cholecystokinin/biosynthesis , Devazepide/pharmacology , Duodenum/drug effects , Duodenum/physiology , Eating/physiology , Fasting , Globulins/pharmacology , Hormone Antagonists/pharmacology , Male , Rats , Rats, Sprague-Dawley , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/metabolism , Satiation/drug effects , Satiation/physiology , Seed Storage Proteins/pharmacology , Soybean Proteins/pharmacology
14.
J Anim Physiol Anim Nutr (Berl) ; 96(2): 214-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21438927

ABSTRACT

Total parenteral nutrition (TPN) results in atrophy of the pancreas, while cholecystokinin (CCK) can significantly stimulate the exocrine pancreas in rodents. This study was designed to examine whether CCK may improve the atrophy of the pancreas in rats on TPN treatment. Forty-eight Sprague-Dawley rats were divided into orally fed and TPN groups and were infused with CCK at a dose of 5 µg/kg/h or the CCK-receptor antagonist devazepide at a dose of 200 µg/kg/h for 10 days. Infusion of CCK caused hypercholecystokininemia (hyperCCKemia) and decreased the atrophy of the pancreas resulting from TPN. The hyperplastic response to CCK in orally fed rats was decreased in the rats given TPN. Devazepide did not influence the pancreatic variables. This study further confirmed that CCK stimulates the exocrine pancreas and decreases the atrophy of the exocrine pancreas resulting from TPN. Our present findings suggest that the trophic effect of CCK on the exocrine pancreas declines in TPN.


Subject(s)
Pancreas/drug effects , Parenteral Nutrition, Total/adverse effects , Sincalide/analogs & derivatives , Animals , Devazepide/pharmacology , Hormone Antagonists/pharmacology , Male , Nootropic Agents/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cholecystokinin/antagonists & inhibitors , Sincalide/pharmacology
15.
Am J Physiol Regul Integr Comp Physiol ; 300(5): R1175-84, 2011 May.
Article in English | MEDLINE | ID: mdl-21325647

ABSTRACT

We hypothesized that protein source in the nutritionally adequate AIN-93G diets fed during gestation, lactation, and weaning influences food intake (FI) regulation in male offspring of Wistar rats. Pregnant rats were fed the recommended casein-based (C) or soy protein-based (S) diet during gestation (experiment 1) or during gestation and lactation (experiment 2). Pups (n = 12 per group) weaned to C or S diets were followed for 9 wk (experiment 1) or 14 wk (experiment 2). At termination, body weight was 5.4% and 9.4% higher, respectively, in offspring of dams fed the S diet. Altered FI regulation was shown by failure of devazepide (a CCK-A receptor blocker) to block FI reduction after protein preloads in offspring of S diet-fed dams, whereas it had a strong effect on offspring of C diet-fed dams (P < 0.005). Similarly, naloxone (an opioid receptor blocker) blocked FI reduction more after casein than after soy protein preloads (P < 0.01). In experiment 2, offspring of dams fed the S diet had higher hypothalamic gene expression of agouti related protein at weaning (P < 0.05), and higher FI was found throughout postweaning (P < 0.0001). FI reduction after protein preloads at week 7 and after glucose preloads at week 13 was greater in offspring of C diet-fed dams (P < 0.05). Plasma insulin at weaning and insulin, ghrelin, and glucagon-like peptide-1 at week 15 were higher in offspring of S diet-fed dams (all P < 0.05). In conclusion, nutritionally complete C and S diets consumed during gestation and lactation differ in their effects on body weight and FI regulation in the offspring. Extending the diet from gestation alone to throughout gestation and lactation exaggerated the adverse effects of the S diet. However, the diet consumed postweaning had little effect on the outcome.


Subject(s)
Animal Nutritional Physiological Phenomena , Appetite Regulation , Behavior, Animal , Caseins/administration & dosage , Dietary Proteins/administration & dosage , Lactation , Maternal Nutritional Physiological Phenomena , Prenatal Exposure Delayed Effects , Soybean Proteins/administration & dosage , Agouti-Related Protein/genetics , Animal Feed , Animals , Animals, Newborn , Appetite Regulation/drug effects , Behavior, Animal/drug effects , Body Weight , Caseins/metabolism , Devazepide/pharmacology , Dietary Proteins/metabolism , Female , Gestational Age , Ghrelin/blood , Glucagon-Like Peptide 1/blood , Hormone Antagonists/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Insulin/blood , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Pregnancy , Rats , Rats, Wistar , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin A/metabolism , Soybean Proteins/metabolism , Time Factors , Weaning
16.
Am J Physiol Regul Integr Comp Physiol ; 301(5): R1557-68, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21865540

ABSTRACT

The discovery that cells in the gastrointestinal (GI) tract express the same molecular receptors and intracellular signaling components known to be involved in taste has generated great interest in potential functions of such post-oral "taste" receptors in the control of food intake. To determine whether taste cues in the GI tract are detected and can directly influence behavior, the present study used a microbehavioral analysis of intake, in which rats drank from lickometers that were programmed to simultaneously deliver a brief yoked infusion of a taste stimulus to the intestines. Specifically, in daily 30-min sessions, thirsty rats with indwelling intraduodenal catheters were trained to drink hypotonic (0.12 M) sodium chloride (NaCl) and simultaneously self-infuse a 0.12 M NaCl solution. Once trained, in a subsequent series of intestinal taste probe trials, rats reduced licking during a 6-min infusion period, when a bitter stimulus denatonium benzoate (DB; 10 mM) was added to the NaCl vehicle for infusion, apparently conditioning a mild taste aversion. Presentation of the DB in isomolar lithium chloride (LiCl) for intestinal infusions accelerated the development of the response across trials and strengthened the temporal resolution of the early licking suppression in response to the arrival of the DB in the intestine. In an experiment to evaluate whether CCK is involved as a paracrine signal in transducing the intestinal taste of DB, the CCK-1R antagonist devazepide partially blocked the response to intestinal DB. In contrast to their ability to detect and avoid the bitter taste in the intestine, rats did not modify their licking to saccharin intraduodenal probe infusions. The intestinal taste aversion paradigm developed here provides a sensitive and effective protocol for evaluating which tastants-and concentrations of tastants-in the lumen of the gut can control ingestion.


Subject(s)
Appetite Regulation/drug effects , Behavior, Animal/drug effects , Cues , Duodenum/innervation , Eating/drug effects , Quaternary Ammonium Compounds/administration & dosage , Receptors, G-Protein-Coupled/drug effects , Taste/drug effects , Afferent Pathways/drug effects , Afferent Pathways/metabolism , Animals , Cholecystokinin/metabolism , Conditioning, Psychological/drug effects , Devazepide/administration & dosage , Hormone Antagonists/administration & dosage , Intubation, Gastrointestinal , Male , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin A/metabolism , Receptors, G-Protein-Coupled/metabolism , Saccharin/administration & dosage , Signal Transduction/drug effects , Sweetening Agents/administration & dosage , Time Factors
17.
Exp Physiol ; 96(4): 439-50, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21239462

ABSTRACT

Cholecystokinin (CCK) provides a meal-related signal that activates brainstem neurons, which have reciprocal interconnections with the hypothalamic paraventricular nucleus. Neurons that express corticotrophin-releasing factor (CRF) in the hypothalamus possess anorexigenic effects and are activated during endotoxaemia. This study investigated the effects of CCK(1) receptor blockade on lipopolysaccharide (LPS)-induced hypophagia and hypothalamic CRF neuronal activation. Male Wistar rats were pretreated with a specific CCK(1) receptor antagonist (devazepide; 1 mg kg(-1); i.p.) or vehicle; 30 min later they received LPS (100 µg kg(-1); i.p.) or saline injection. Food intake, corticosterone responses and Fos-CRF and Fos-α-melanocyte-stimulating hormone (α-MSH) immunoreactivity in the hypothalamus and Fos-tyrosine hydroxylase immunoreactivity in the nucleus of the solitary tract (NTS) were evaluated. In comparison with saline treatment, LPS administration decreased food intake and increased plasma corticosterone levels, as well as the number of Fos-CRF and Fos- tyrosine hydroxylase double-labelled neurons in vehicle-pretreated rats; no change in Fos-α-MSH immunoreactivity was observed after LPS injection. In saline-treated animals, devazepide pretreatment increased food intake, but it did not modify other parameters compared with vehicle-pretreated rats. Devazepide pretreatment partly reversed LPS-induced hypophagia and Fos-CRF and brainstem neuronal activation. Devazepide did not modify the corticosterone and Fos-α-MSH responses in rats treated with LPS. In conclusion, the present data suggest that LPS-induced hypophagia is mediated at least in part by CCK effects, via CCK(1) receptor, on NTS and hypothalamic CRF neurons.


Subject(s)
Cholecystokinin/metabolism , Corticotropin-Releasing Hormone/metabolism , Endotoxins/pharmacology , Hyperphagia/metabolism , Hypothalamus/metabolism , Pituitary Hormone-Releasing Hormones/metabolism , Animals , Brain Stem/metabolism , Corticosterone/blood , Devazepide/pharmacology , Eating/drug effects , Endotoxemia/chemically induced , Endotoxemia/metabolism , Hyperphagia/chemically induced , Lipopolysaccharides , Male , Neurons/enzymology , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin A/metabolism , Solitary Nucleus/metabolism , Tyrosine 3-Monooxygenase/metabolism , alpha-MSH/metabolism
18.
Neurochem Res ; 36(1): 129-38, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20953702

ABSTRACT

Electroacupuncture (EA) has been successfully used to alleviate pain produced by various noxious stimulus. Cholecystokinin-8 (CCK-8) is a neuropeptide involved in the mediation of pain. We have previously shown that CCK-8 could antagonize the analgesic effects of EA on pain-excited neurons (PENs) and pain-inhibited neurons (PINs) in the nucleus parafascicularis (nPf). However, its mechanism of action is not clear. In the present study, we applied behavioral and neuroelectrophysiological methods to determine whether the mechanisms of CCK-8 antagonism to EA analgesia are mediated through the CCK-A receptors of PENs and PINs in the nPf of rats. We found that focusing radiant heat on the tail of rats caused a simultaneous increase in the evoked discharge of PENs or a decrease in the evoked discharge of PINs in the nPf and the tail-flick reflex. This showed that radiant heat could induce pain. EA stimulation at the bilateral ST 36 acupoints in rats for 15 min resulted in an inhibition of the electrical activity of PEN, potentiation of the electrical activity of PIN, and prolongation in tail-flick latency (TFL), i.e. EA stimulation produced an analgesic effect. The analgesic effect of EA was antagonized when CCK-8 was injected into the intracerebral ventricle of rats. The antagonistic effect of CCK-8 on EA analgesia was reversed by an injection of CCK-A receptor antagonist L-364,718 (100 ng/µl) into the nPf of rats. Our results suggest that the pain-related neurons in the nPf have an important role in mediating EA analgesia. L-364,718 potentiates EA analgesia through the CCK-A receptor of PENs and PINs in the nPf.


Subject(s)
Acupuncture Analgesia/methods , Devazepide/pharmacology , Electroacupuncture/methods , Intralaminar Thalamic Nuclei/cytology , Pain Management , Receptor, Cholecystokinin A/metabolism , Sensory Receptor Cells/drug effects , Animals , Cholecystokinin/metabolism , Hormone Antagonists/pharmacology , Male , Pain Measurement , Peptide Fragments/metabolism , Random Allocation , Rats , Rats, Wistar , Receptor, Cholecystokinin A/antagonists & inhibitors , Sensory Receptor Cells/metabolism
19.
Physiol Res ; 70(4): 579-590, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34062082

ABSTRACT

Prolactin-releasing peptide (PrRP) has been proposed to mediate the central satiating effects of cholecystokinin (CCK) through the vagal CCK1 receptor. PrRP acts as an endogenous ligand of G protein-coupled receptor 10 (GPR10), which is expressed at the highest levels in brain areas related to food intake regulation, e.g., the paraventricular hypothalamic nucleus (PVN) and nucleus of the solitary tract (NTS). The NTS and PVN are also significantly activated after peripheral CCK administration. The aim of this study was to determine whether the endogenous PrRP neuronal system in the brain is involved in the central anorexigenic effect of the peripherally administered CCK agonist JMV236 or the CCK1 antagonist devazepide and whether the CCK system is involved in the central anorexigenic effect of the peripherally applied lipidized PrRP analog palm-PrRP31 in fasted lean mice. The effect of devazepide and JMV236 on the anorexigenic effects of palm-PrRP31 as well as devazepide combined with JMV236 and palm-PrRP31 on food intake and Fos cell activation in the PVN and caudal NTS was examined. Our results suggest that the anorexigenic effect of JMV236 is accompanied by activation of PrRP neurons of the NTS in a CCK1 receptor-dependent manner. Moreover, while the anorexigenic effect of palm-PrRP31 was not affected by JMV236, it was partially attenuated by devazepide in fasted mice. The present findings indicate that the exogenously influenced CCK system may be involved in the central anorexigenic effect of peripherally applied palm-PrRP31, which possibly indicates some interaction between the CCK and PrRP neuronal systems.


Subject(s)
Appetite Depressants/administration & dosage , Cholecystokinin/metabolism , Eating/drug effects , Feeding Behavior/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Prolactin-Releasing Hormone/analogs & derivatives , Solitary Nucleus/drug effects , Animals , Chemokines, CC/drug effects , Chemokines, CC/metabolism , Devazepide/administration & dosage , Fasting , Hormone Antagonists/administration & dosage , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Mice, Inbred C57BL , Paraventricular Hypothalamic Nucleus/metabolism , Peptide Fragments/administration & dosage , Prolactin-Releasing Hormone/administration & dosage , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction , Sincalide/administration & dosage , Sincalide/analogs & derivatives , Solitary Nucleus/metabolism
20.
Mol Pain ; 6: 72, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-20979661

ABSTRACT

BACKGROUND: Z-360 is an orally active cholecystokinin-2 (CCK2)/gastrin receptor antagonist currently under development as a therapeutic drug for pancreatic cancer. It was previously reported that Z-360 treatment in combination with gemcitabine prolonged the survival period in a lethal pancreatic cancer xenograft model in mice. In a phase Ib/IIa clinical study, Z-360 treatment displayed a trend of reduced pain in patients with advanced pancreatic cancer in combination with gemcitabine including analgesics such as opioids. Here, we investigated the mechanism of analgesic action of Z-360 in a severe cancer-induced pain model in mice, which is considered to be opioid-resistant, by examining ephrin B1 gene expression, N-methyl-D-aspartate receptor NR2B subunit phosphorylation, and interleukin-1ß (IL-1ß) production. RESULTS: In a mouse model of cancer-induced pain, ephrin B1 gene expression in dorsal root ganglia (DRGs) and the phosphorylation of NR2B in the spinal cord were induced. Z-360 treatment inhibited both ephrin B1 gene expression and the phosphorylation of NR2B. In addition, IL-1ß production increased in the cancer-inoculated hind paw of mice, but could be suppressed by treatment with Z-360. Moreover, we observed that the CCK1 receptor antagonist devazepide similarly suppressed up-regulation of ephrin B1 gene expression and IL-1ß production, and that the intraperitoneal injection of sulfated CCK-8 induced the production of IL-1ß in the cancer-inoculated region. CONCLUSIONS: We have identified a novel pain cascade, in which IL-1ß production in cancer-inoculated regions induces ephrin B1 gene expression in DRGs and then ephrin B1 enhances the tyrosine phosphorylation of NR2B via Eph B receptor in the spinal cord. Notably, Z-360 relieves cancer-induced pain by preventing this pain cascade through the suppression of IL-1ß production, likely via the blockade of CCK1 receptor. The pre-clinical results presented here support the analgesic action of Z-360 in pancreatic cancer patients with severe, opioid-resistant pain. Pre-clinical and clinical results have demonstrated that Z-360 combined with gemcitabine represents a promising pancreatic cancer therapy approach with characteristic analgesic effects in addition to the prolongation of survival.


Subject(s)
Benzodiazepinones/therapeutic use , Ephrin-B1/genetics , Interleukin-1beta/biosynthesis , Pain/etiology , Pancreatic Neoplasms/drug therapy , Receptors, N-Methyl-D-Aspartate/metabolism , Up-Regulation , Animals , Benzodiazepinones/pharmacology , Cell Line, Tumor , Devazepide/pharmacology , Disease Models, Animal , Ephrin-B1/metabolism , Extremities , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Expression Regulation, Neoplastic/drug effects , Injections , Interleukin-1beta/administration & dosage , Interleukin-1beta/pharmacology , Mice , Pain/genetics , Pain/pathology , Pancreatic Neoplasms/complications , Phosphorylation/drug effects , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Sincalide/analogs & derivatives , Sincalide/pharmacology , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL