Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Biol ; 22(7): e3002673, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39083706

ABSTRACT

Development of optimal therapeutics for disease states that can be associated with increased membrane cholesterol requires better molecular understanding of lipid modulation of the drug target. Type 1 cholecystokinin receptor (CCK1R) agonist actions are affected by increased membrane cholesterol, enhancing ligand binding and reducing calcium signaling, while agonist actions of the closely related CCK2R are not. In this work, we identified a set of chimeric human CCK1R/CCK2R mutations that exchange the cholesterol sensitivity of these 2 receptors, providing powerful tools when expressed in CHO and HEK-293 model cell lines to explore mechanisms. Static, low energy, high-resolution structures of the mutant CCK1R constructs, stabilized in complex with G protein, were not substantially different, suggesting that alterations to receptor dynamics were key to altered function. We reveal that cholesterol-dependent dynamic changes in the conformation of the helical bundle of CCK receptors affects both ligand binding at the extracellular surface and G protein coupling at the cytosolic surface, as well as their interrelationships involved in stimulus-response coupling. This provides an ideal setting for potential allosteric modulators to correct the negative impact of membrane cholesterol on CCK1R.


Subject(s)
Cholesterol , GTP-Binding Proteins , Protein Binding , Receptor, Cholecystokinin A , Receptor, Cholecystokinin B , Animals , Humans , CHO Cells , Cholesterol/metabolism , Cricetulus , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/genetics , HEK293 Cells , Ligands , Mutation , Protein Conformation , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin B/metabolism , Receptor, Cholecystokinin B/genetics
2.
Exp Eye Res ; 188: 107763, 2019 11.
Article in English | MEDLINE | ID: mdl-31421135

ABSTRACT

Primary open-angle glaucoma (POAG) is a leading cause of irreversible blindness, and individuals with ocular hypertension are at risk to develop POAG. Currently, the only modifiable risk factor for glaucoma progression is lowering of intraocular pressure (IOP). A novel mechanism for lowering IOP involves activation of the type B natriuretic peptide receptor (NPR-B), the naturally occurring agonist of which is C-type natriuretic peptide (CNP). Being a cyclic peptide of 22 amino acids, CNP does not readily penetrate the cornea and its ocular hypotensive effect requires intraocular injection. TAK-639 is a synthetic, cornea-permeable, 9-amino acid CNP analog has been studied for the treatment of ocular hypertension and POAG. We assessed TAK-639 in a receptor binding profile and the effects of TAK-639 on NPR-B-mediated cyclic GMP production in cultured transformed human trabecular meshwork (TM) cells (GTM-3). We also evaluated the effects of topical ocular administration of TAK-639 on mouse IOP and aqueous humor dynamics. Among 89 non-natriuretic peptide receptors, transporters, and channels evaluated, TAK-639 at 10 µM displaced ligand binding by more than 50% to only two receptors: the type 2 angiotensin receptor (IC50 = 8.2 µM) and the cholecystokinin A receptor (IC50 = 25.8 µM). In vitro, TAK-639 selectively activates NPR-B (EC50 = 61 ±â€¯11 nM; GTM-3 cells) relative to NPR-A (EC50 = 2179 ±â€¯670 nM; 293T cells). In vivo, TAK-639 lowered mouse IOP by three mechanisms: increase in aqueous humor outflow facility (C), reduction in the aqueous humor formation rate (Fin), and reduction in episcleral venous pressure (Pe). The maximum mean IOP decreases from baseline were -12.1%, -21.0%, and -36.1% for 0.1%, 0.3%, and 0.6% doses of TAK-639, respectively. Maximum IOP-lowering effect was seen at 2 h, and the duration of action was >6 h. With TAK-639 0.6%, at 2 h post-dose, aqueous outflow facility (C) increased by 155.8%, Fin decreased by 41.0%, the uveoscleral outflow rate (Fu) decreased by 52.6%, and Pe decreased by 31.5% (all p < 0.05). No ocular adverse effects were observed. TAK-639 is an efficacious IOP-lowering agent, with a unique combination of mechanisms of action on both aqueous formation and aqueous outflow facility. Further study of this mechanism of treatment may optimize pharmacologic outcomes and provide disease management in patients with POAG and ocular hypertension.


Subject(s)
Aqueous Humor/physiology , Intraocular Pressure/drug effects , Natriuretic Peptide, C-Type/analogs & derivatives , Natriuretic Peptide, C-Type/pharmacology , Trabecular Meshwork/drug effects , Administration, Ophthalmic , Animals , Cell Line, Transformed , Cyclic GMP/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Ophthalmic Solutions , Receptor, Angiotensin, Type 2/metabolism , Receptor, Cholecystokinin A/metabolism , Receptors, Atrial Natriuretic Factor/metabolism , Tonometry, Ocular , Trabecular Meshwork/metabolism
3.
Biochim Biophys Acta Mol Cell Res ; 1864(7): 1153-1164, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28288880

ABSTRACT

The orexin (OX1R) and cholecystokinin A (CCK1R) receptors play opposing roles in the migration of the human colon cancer cell line HT-29, and may be involved in the pathogenesis and pathophysiology of cancer cell invasion and metastasis. OX1R and CCK1R belong to family A of the G-protein-coupled receptors (GPCRs), but the detailed mechanisms underlying their functions in solid tumor development remain unclear. In this study, we investigated whether these two receptors heterodimerize, and the results revealed novel signal transduction mechanisms. Bioluminescence and Förster resonance energy transfer, as well as proximity ligation assays, demonstrated that OX1R and CCK1R heterodimerize in HEK293 and HT-29 cells, and that peptides corresponding to transmembrane domain 5 of OX1R impaired heterodimer formation. Stimulation of OX1R and CCK1R heterodimers with both orexin-A and CCK decreased the activation of Gαq, Gαi2, Gα12, and Gα13 and the migration of HT-29 cells in comparison with stimulation with orexin-A or CCK alone, but did not alter GPCR interactions with ß-arrestins. These results suggest that OX1R and CCK1R heterodimerization plays an anti-migratory role in human colon cancer cells.


Subject(s)
Cell Movement , GTP-Binding Protein alpha Subunits/metabolism , Orexin Receptors/metabolism , Protein Multimerization , Receptor, Cholecystokinin A/metabolism , Signal Transduction , HEK293 Cells , HT29 Cells , Humans , Orexin Receptors/genetics , Protein Binding , Protein Domains , Receptor, Cholecystokinin A/genetics , beta-Arrestins/metabolism
4.
Am J Physiol Renal Physiol ; 313(1): F20-F29, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28298361

ABSTRACT

The natriuretic hormone CCK exhibits its gene transcripts in total kidney extracts. To test the possibility of CCK acting as an intrarenal mediator of sodium excretion, we examined mouse kidneys by 1) an in situ hybridization technique for CCK mRNA in animals fed a normal- or a high-sodium diet; 2) immuno-electron microscopy for the CCK peptide, 3) an in situ hybridization method and immunohistochemistry for the CCK-specific receptor CCKAR; 4) confocal image analysis of receptor-mediated Ca2+ responses in isolated renal tubules; and 5) metabolic cage experiments for the measurement of urinary sodium excretion in high-salt-fed mice either treated or untreated with the CCKAR antagonist lorglumide. Results showed the CCK gene to be expressed intensely in the inner medulla and moderately in the inner stripe of the outer medulla, with the expression in the latter being enhanced by high sodium intake. Immunoreactivity for the CCK peptide was localized to the rough endoplasmic reticulum of the medullary interstitial cells in corresponding renal regions, confirming it to be a secretory protein. Gene transcripts, protein products, and the functional activity for CCKAR were consistently localized to the late proximal tubule segments (S2 and S3) in the medullary rays, and the outer stripe of the outer medulla. Lorglumide significantly diminished natriuretic responses of mice to a dietary sodium load without altering the glomerular filtration rate. These findings suggest that the medullary interstitial cells respond to body fluid expansion by CCK release for feedback regulation of the late proximal tubular reabsorption.


Subject(s)
Cholecystokinin/metabolism , Kidney Medulla/metabolism , Kidney Tubules, Proximal/metabolism , Natriuresis , Signal Transduction , Sodium, Dietary/administration & dosage , Water-Electrolyte Balance , Animals , Calcium/metabolism , Cholecystokinin/antagonists & inhibitors , Cholecystokinin/genetics , Feedback, Physiological , Hormone Antagonists/pharmacology , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kidney Medulla/drug effects , Kidney Medulla/ultrastructure , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/ultrastructure , Male , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Immunoelectron , Natriuresis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/metabolism , Signal Transduction/drug effects , Time Factors , Water-Electrolyte Balance/drug effects
5.
Tumour Biol ; 37(4): 4579-84, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26508021

ABSTRACT

Cholecystokinin and gastrin receptors are upregulated in many human digestive malignancies; however, the correlation of their expressions with severity of colon carcinoma remains sketchy. Here, we determined the expression of cholecystokinin-1 and cholecystokinin-2 receptor, CCK1R and CCK2R, in colon carcinomas and investigated their correlations with clinicopathological characteristics and 1-year survival rate. Expression of CCK1R and CCK2R was determined by immunohistochemical assay in tissue samples obtained from 97 surgical specimens. Clinicopathological character analysis revealed that higher expression of cytoplasmic CCK1R and CCK2R was significantly associated with several variables including the depth of tumor invasion (P = 0.001), venous invasion (P = 0.023), and progression stage (P = 0.013). In addition, immunohistochemical staining revealed statistically significant associations of nuclear CCK1R expression with higher lymphatic invasion (P = 0.042), progression stage (P = 0.025), and unfavorable survival (P = 0.025). Interestingly, we found no link between nuclear CCK2R expression and all the clinicopathological characteristics examined. Taken these, our findings indicate that nuclear CCK1R represents a potential biomarker for poor prognosis, and CCK1R may play a role differing from CCK2R in colon carcinogenesis.


Subject(s)
Biomarkers, Tumor/metabolism , Colonic Neoplasms/metabolism , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin B/metabolism , Aged , Aged, 80 and over , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Survival Analysis , Taiwan
6.
Horm Behav ; 78: 79-85, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26522495

ABSTRACT

The current study tested the hypothesis that cholecystokinin (CCK) A receptor (CCKAR) in areas supplied by the celiac artery (CA), stomach and upper duodenum, and the cranial mesenteric artery (CMA), small and parts of the large intestine, is necessary for reduction of meal size, prolongation of the intermeal interval (time between first and second meal) and increased satiety ratio (intermeal interval/meal size or amount of food consumed during any given unit of time) by the non-nutrient stimulator of endogenous CCK release camostat. Consistent with our previous findings camostat reduced meal size, prolonged the intermeal interval and increased the satiety ratio. Here, we report that blocking CCKAR in the area supplied by the celiac artery attenuated reduction of meal size by camostat more so than the cranial mesenteric artery route. Blocking CCKAR in the area supplied by the cranial mesenteric artery attenuated prolongation of the intermeal interval length and increased satiety ratio by camostat more so than the celiac artery route. Blocking CCKAR in the areas supplied by the femoral artery (control) failed to alter the feeding responses evoked by camostat. These results support the hypothesis that CCKAR in the area supplied by the CA is necessary for reduction of meal size by camostat whereas CCKAR in the area supplied by the CMA is necessary for prolongation of the intermeal interval and increased satiety ratio by this substance. Our results demonstrate that meal size and intermeal interval length by camostat are regulated through different gastrointestinal sites.


Subject(s)
Celiac Artery/metabolism , Cholecystokinin/metabolism , Eating/physiology , Feeding Behavior/physiology , Gabexate/analogs & derivatives , Mesenteric Artery, Superior/metabolism , Protease Inhibitors/pharmacology , Receptor, Cholecystokinin A/metabolism , Animals , Eating/drug effects , Esters , Feeding Behavior/drug effects , Femoral Artery/metabolism , Gabexate/administration & dosage , Gabexate/pharmacology , Guanidines , Male , Protease Inhibitors/administration & dosage , Rats , Rats, Sprague-Dawley , Time Factors
7.
Biochem Genet ; 54(5): 665-75, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27287528

ABSTRACT

In the present study, we investigated expression pattern of Cholecystokinin type A receptor (CCKAR) in relation to its commonly studied polymorphism (rs1800857, T/C) in gallstone disease (GSD) patients and controls. A total of 502 subjects (272 GSD and 230 controls) were enrolled, and genotyping was performed by evaluating restriction fragments of PstI digested DNA. For analyzing expression pattern of CCKAR in relation to polymorphism, gallbladder tissue samples from 80 subjects (GSD-55; control-25) were studied. Expression of CCKAR mRNA was evaluated by reverse transcriptase-PCR and confirmed using real-time PCR. Protein expression was evaluated by enzyme-linked immunosorbent assay. We observed significantly (p < 0.0001) lower expression of CCKAR mRNA and protein in GSD tissues as compared with control. Significantly higher frequency of A1/A1 genotype (C/T transition) (p = 0.0005) was observed for GSD as compared with control. Expression of CCKAR protein was found to be significantly lower (p < 0.0001) in A1/A1 genotype as compared with other genotypes for GSD patients. Perhaps, this is the first report providing evidence of alteration in CCKAR expression in relation to its polymorphism elucidating the molecular pathway of the disease. Additional investigations with lager sample size are needed to confirm these findings.


Subject(s)
Gallstones/genetics , Polymorphism, Single Nucleotide , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/metabolism , Adult , Disease Susceptibility , Down-Regulation , Female , Gallstones/metabolism , Genotyping Techniques , Humans , India , Male , Middle Aged , Tissue Distribution , Young Adult
8.
Mol Pharmacol ; 87(1): 130-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25319540

ABSTRACT

Understanding the molecular basis of ligand binding to receptors provides insights useful for rational drug design. This work describes development of a new antagonist radioligand of the type 1 cholecystokinin receptor (CCK1R), (2-fluorophenyl)-2,3-dihydro-3-[(3-isoquinolinylcarbonyl)amino]-6-methoxy-2-oxo-l-H-indole-3-propanoate (T-0632), and exploration of the molecular basis of its binding. This radioligand bound specifically with high affinity within an allosteric pocket of CCK1R. T-0632 fully inhibited binding and action of CCK at this receptor, while exhibiting no saturable binding to the closely related type 2 cholecystokinin receptor (CCK2R). Chimeric CCK1R/CCK2R constructs were used to explore the molecular basis of T-0632 binding. Exchanging exonic regions revealed the functional importance of CCK1R exon 3, extending from the bottom of transmembrane segment (TM) 3 to the top of TM5, including portions of the intramembranous pocket as well as the second extracellular loop region (ECL2). However, CCK1R mutants in which each residue facing the pocket was changed to that present in CCK2R had no negative impact on T-0632 binding. Extending the chimeric approach to ECL2 established the importance of its C-terminal region, and site-directed mutagenesis of each nonconserved residue in this region revealed the importance of Ser(208) at the top of TM5. A molecular model of T-0632-occupied CCK1R was consistent with these experimental determinants, also identifying Met(121) in TM3 and Arg(336) in TM6 as important. Although these residues are conserved in CCK2R, mutating them had a distinct impact on the two closely related receptors, suggesting differential orientation. This establishes the molecular basis of binding of a highly selective nonpeptidyl allosteric antagonist of CCK1R, illustrating differences in docking that extend beyond determinants attributable to distinct residues lining the intramembranous pocket in the two receptor subtypes.


Subject(s)
Amino Acids/metabolism , Indoles/chemistry , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/metabolism , Animals , Binding Sites/drug effects , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetulus , Humans , Indoles/pharmacology , Molecular Docking Simulation , Mutagenesis, Site-Directed , Radioligand Assay , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism
9.
J Biol Chem ; 289(26): 18314-26, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24825903

ABSTRACT

Cholecystokinin (CCK) stimulates the type 1 CCK receptor (CCK1R) to elicit satiety after a meal. Agonists with this activity, although potentially useful for treatment of obesity, can also have side effects and toxicities of concern, making the development of an intrinsically inactive positive allosteric modulator quite attractive. Positive allosteric modulators also have the potential to correct the defective receptor-G protein coupling observed in the high membrane cholesterol environment described in metabolic syndrome. Current model systems to study CCK1R in such an environment are unstable and expensive to maintain. We now report that the Y140A mutation within a cholesterol-binding motif and the conserved, class A G protein-coupled receptor-specific (E/D)RY signature sequence results in ligand binding and activity characteristics similar to wild type CCK1R in a high cholesterol environment. This is true for natural CCK, as well as ligands with distinct chemistries and activity profiles. Additionally, the Y140A construct also behaved like CCK1R in high cholesterol in regard to its internalization, sensitivity to a nonhydrolyzable GTP analog, and anisotropy of a bound fluorescent CCK analog. Chimeric CCK1R/CCK2R constructs that systematically changed the residues in the allosteric ligand-binding pocket were studied in the presence of Y140A. This established increased importance of unique residues within TM3 and reduced the importance of TM2 for binding in the presence of this mutation, with the agonist trigger likely pulled away from its Leu(356) target on TM7. The distinct conformation of this intramembranous pocket within Y140A CCK1R provides an opportunity to normalize this by using a small molecule allosteric ligand, thereby providing safe and effective correction of the coupling defect in metabolic syndrome.


Subject(s)
Cholesterol/metabolism , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cholesterol/chemistry , Cricetulus , Humans , Kinetics , Models, Molecular , Mutation, Missense , Protein Binding , Receptor, Cholecystokinin A/chemistry
10.
BMC Genomics ; 16: 431, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-26044654

ABSTRACT

BACKGROUND: Domestic goats (Capra hircus) have been selected to play an essential role in agricultural production systems, since being domesticated from their wild progenitor, bezoar (Capra aegagrus). A detailed understanding of the genetic consequences imparted by the domestication process remains a key goal of evolutionary genomics. RESULTS: We constructed the reference genome of bezoar and sequenced representative breeds of domestic goats to search for genomic changes that likely have accompanied goat domestication and breed formation. Thirteen copy number variation genes associated with coat color were identified in domestic goats, among which ASIP gene duplication contributes to the generation of light coat-color phenotype in domestic goats. Analysis of rapidly evolving genes identified genic changes underlying behavior-related traits, immune response and production-related traits. CONCLUSION: Based on the comparison studies of copy number variation genes and rapidly evolving genes between wild and domestic goat, our findings and methodology shed light on the genetic mechanism of animal domestication and will facilitate future goat breeding.


Subject(s)
Genome , Goats/genetics , Amino Acid Sequence , Animals , Animals, Domestic/genetics , Animals, Wild/genetics , Biological Evolution , Breeding , DNA/analysis , DNA/isolation & purification , DNA Copy Number Variations , Genetic Variation , Immune System/metabolism , Male , Molecular Sequence Data , Myelin and Lymphocyte-Associated Proteolipid Proteins/classification , Myelin and Lymphocyte-Associated Proteolipid Proteins/genetics , Nervous System/metabolism , Phylogeny , Protein Structure, Tertiary , Receptor, Cholecystokinin A/chemistry , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/metabolism , Sequence Alignment
11.
Am J Physiol Gastrointest Liver Physiol ; 309(5): G377-86, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26138469

ABSTRACT

Dysfunction of the type 1 cholecystokinin (CCK) receptor (CCK1R) as a result of increased gallbladder muscularis membrane cholesterol has been implicated in the pathogenesis of cholesterol gallstones. Administration of ursodeoxycholic acid, which is structurally related to cholesterol, has been shown to have beneficial effects on gallstone formation. Our aims were to explore the possible direct effects and mechanism of action of bile acids on CCK receptor function. We studied the effects of structurally related hydrophobic chenodeoxycholic acid and hydrophilic ursodeoxycholic acid in vitro on CCK receptor function in the setting of normal and elevated membrane cholesterol. We also examined their effects on a cholesterol-insensitive CCK1R mutant (Y140A) disrupting a key site of cholesterol action. The results show that, similar to the impact of cholesterol on CCK receptors, bile acid effects were limited to CCK1R, with no effects on CCK2R. Chenodeoxycholic acid had a negative impact on CCK1R function, while ursodeoxycholic acid had no effect on CCK1R function in normal membranes but was protective against the negative impact of elevated cholesterol on this receptor. The cholesterol-insensitive CCK1R mutant Y140A was resistant to effects of both bile acids. These data suggest that bile acids compete with the action of cholesterol on CCK1R, probably by interacting at the same site, although the conformational impact of each bile acid appears to be different, with ursodeoxycholic acid capable of correcting the abnormal conformation of CCK1R in a high-cholesterol environment. This mechanism may contribute to the beneficial effect of ursodeoxycholic acid in reducing cholesterol gallstone formation.


Subject(s)
Receptor, Cholecystokinin A/metabolism , Ursodeoxycholic Acid/pharmacology , Animals , Binding Sites , CHO Cells , Chenodeoxycholic Acid/pharmacology , Cholesterol/pharmacology , Cricetinae , Cricetulus , Digestion/drug effects , Mutation , Protein Binding , Receptor, Cholecystokinin A/genetics
12.
Eur J Immunol ; 44(2): 489-99, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24301797

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are specialized in rapid and massive secretion of type I interferon in response to foreign nuclei acids. Combined with their antigen presentation capacity, this powerful functionality enables pDCs to orchestrate innate and adaptive immune responses. Cholecystokinin octapeptide (CCK8) is a potent immunomodulator, whose role in pDCs function is unknown. In this study, we found that two different cholecystokinin receptors, CCK1R and CCK2R, are expressed on human peripheral blood pDCs. Exogenous CCK8 was able to modulate the TLR-induced activation of pDCs, including phenotypic maturation, IFN-α synthesis and secretion, and could also regulate the potential of pDCs to induce adaptive immune responses in vitro. CCK8 inhibited TLR9-induced activation of tumor-necrosis factor receptor-associated factor 6, which is an important adapter protein in activation of interferon-regulatory factor (IRF)5 and IRF7, possibly through CCK2R, by evoking the activity of protein kinase (PK)A and reducing the activity of PKC. All these results indicate that CCK8 can inhibit the TLR9-induced phenotypic maturation and activation of pDCs, acting through CCK2R by modulating the tumor-necrosis factor receptor-associated factor 6 signaling pathways.


Subject(s)
Dendritic Cells/metabolism , Signal Transduction/genetics , Sincalide/genetics , TNF Receptor-Associated Factor 6/genetics , Toll-Like Receptor 9/genetics , Humans , Interferon-alpha/genetics , Interferon-alpha/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin B/genetics , Receptor, Cholecystokinin B/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Sincalide/metabolism , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptor 9/metabolism
13.
Histochem Cell Biol ; 143(3): 301-12, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25249350

ABSTRACT

Information concerning the cellular localization of cholecystokinin (CCK)-1 receptors has been discrepant and remained scanty at ultrastructural levels. The present immunohistochemical study at light and electron microscopic levels revealed the distinct localization of CCK1 receptors in visceral organs. Immunohistochemistry by use of a purified antibody against mouse CCK1 receptor was applied to fixed tissue sections of the pancreas, gallbladder, stomach, and intestine of mice. A silver-intensified immunogold method revealed the subcellular localization under electron microscope. The immunoreactivity for CCK1 receptors was selectively found in the basolateral membrane of pancreatic acinar cells and gastric chief cells but was absent in pancreatic islets and gastric D cells. Another intense expression in the gut was seen in the myenteric nerve plexus of the antro-duodenal region and some populations of c-Kit-expressing pacemaker cells in the duodenal musculature. The gallbladder contained smooth muscle fibers with an intense immunoreactivity of CCK1 receptors on cell surfaces. The restricted localization of CCK1 receptors on the basolateral membrane of pancreatic acinar cells and gastric chief cells, along with their absence in the islets of Langerhans and gastric D cells, provides definitive information concerning the regulatory mechanism by circulating CCK. Especially, the subcellular localization in the acinar cells completes the investigation for the detection of circulating CCK by the basolateral membrane.


Subject(s)
Gallbladder/cytology , Pancreas/cytology , Receptor, Cholecystokinin A/analysis , Receptor, Cholecystokinin A/metabolism , Stomach/cytology , Animals , Gallbladder/ultrastructure , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred Strains , Microscopy, Electron , Molecular Sequence Data , Pancreas/ultrastructure , Receptor, Cholecystokinin A/ultrastructure , Stomach/ultrastructure
14.
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
15.
Biosci Biotechnol Biochem ; 79(7): 1164-71, 2015.
Article in English | MEDLINE | ID: mdl-25765851

ABSTRACT

We investigated the effect of daidzein feeding and estradiol treatment on food intake in cholecystokinin-1 receptor (CCK1R) deficiency, leptin receptor (ObRb) deficiency rats and their wild-type rats. These rats underwent an ovariectomy or a sham operation. For the 5 week experiment, each rat was divided in three groups: control, daidzein (150 mg/kg diet), and estradiol (4.2 µg/rat/day) groups. In both CCK1R+ and CCK1R- rats, daidzein feeding and estradiol treatment significantly decreased food intake. Daidzein feeding significantly reduced food intake in ovariectomized ObRb- rats, although not in ObRb+ rats. Estradiol treatment significantly lowered food intake in ovariectomized ObRb+ and ObRb- rats. In the ovariectomized rats, estradiol treatment significantly increases uterine weight, while daidzein feeding did not change it, suggesting that daidzein might have no or weak estrogenic effect in our experiment. These results suggest that CCK1R and ObRb signalings were not essential for the daidzein- and estradiol-induced anorectic action.


Subject(s)
Appetite Depressants/pharmacology , Estradiol/pharmacology , Isoflavones/pharmacology , Receptor, Cholecystokinin A/genetics , Receptors, Leptin/metabolism , Animals , Body Weight/drug effects , Eating/drug effects , Estradiol/blood , Female , Organ Size/drug effects , Ovariectomy , Rats, Long-Evans , Rats, Mutant Strains , Receptor, Cholecystokinin A/metabolism , Receptors, Leptin/genetics , Uterus/drug effects
16.
J Physiol ; 592(7): 1705-20, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24492842

ABSTRACT

The gut hormone cholecystokinin (CCK) acts at subdiaphragmatic vagal afferents to induce renal and splanchnic sympathoinhibition and vasodilatation, via reflex inhibition of a subclass of cardiovascular-controlling neurons in the rostroventrolateral medulla (RVLM). These sympathoinhibitory and vasodilator responses are blunted in obese, hypertensive rats and our aim in the present study was to determine whether this is attributable to (i) altered sensitivity of presympathetic vasomotor RVLM neurons, and (ii) aberrant peripheral or central signalling mechanisms. Using a diet-induced obesity model, male Sprague-Dawley rats exhibited either an obesity-prone (OP) or obesity-resistant (OR) phenotype when placed on a medium high fat diet for 13-15 weeks; control animals were placed on a low fat diet. OP animals had elevated resting arterial pressure compared to OR/control animals (P < 0.05). Barosensitivity of RVLM neurons was significantly attenuated in OP animals (P < 0.05), suggesting altered baroreflex gain. CCK induced inhibitory responses in RVLM neurons of OR/control animals but not OP animals. Subdiaphragmatic vagal nerve responsiveness to CCK and CCK1 receptor mRNA expression in nodose ganglia did not differ between the groups, but CCK induced significantly less Fos-like immunoreactivity in both the nucleus of the solitary tract and the caudal ventrolateral medulla of OP animals compared to controls (P < 0.05). These results suggest that blunted sympathoinhibitory and vasodilator responses in obesity-related hypertension are due to alterations in RVLM neuronal responses, resulting from aberrant central but not peripheral signalling mechanisms. In obesity, blunted sympathoinhibitory mechanisms may lead to increased regional vascular resistance and contribute to the development of hypertension.


Subject(s)
Baroreflex , Hypertension/etiology , Medulla Oblongata/physiopathology , Neural Inhibition , Obesity/complications , Signal Transduction , Sympathetic Nervous System/physiopathology , Animals , Arterial Pressure , Cholecystokinin/pharmacology , Disease Models, Animal , Hypertension/metabolism , Hypertension/physiopathology , Male , Medulla Oblongata/drug effects , Medulla Oblongata/metabolism , Neural Inhibition/drug effects , Nodose Ganglion/metabolism , Nodose Ganglion/physiopathology , Obesity/metabolism , Obesity/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Receptor, Cholecystokinin A/genetics , Receptor, Cholecystokinin A/metabolism , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
17.
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
18.
Diabetologia ; 56(6): 1425-35, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23462797

ABSTRACT

AIMS/HYPOTHESIS: Cholecystokinin (CCK) and leptin are important hormones with effects on energy balance. The present study assessed the biological effects of (pGlu-Gln)-CCK-8 and [D-Leu-4]-OB3, smaller isoforms of CCK and leptin, respectively. METHODS: The actions and overall therapeutic use of (pGlu-Gln)-CCK-8 and [D-Leu-4]-OB3, alone and in combination, were evaluated in normal and high-fat-fed mice. RESULTS: (pGlu-Gln)-CCK-8 had prominent (p < 0.01 to p < 0.001), acute feeding-suppressive effects, which were significantly augmented (p < 0.05 to p < 0.01) by [D-Leu-4]-OB3. In agreement, the acute dose-dependent glucose-lowering and insulinotropic actions of (pGlu-Gln)-CCK-8 were significantly enhanced by concurrent administration of [D-Leu-4]-OB3. Twice daily injection of (pGlu-Gln)-CCK-8 alone and in combination with [D-Leu-4]-OB3 in high-fat-fed mice for 18 days decreased body weight (p < 0.05 to p < 0.001), energy intake (p < 0.01), circulating triacylglycerol (p < 0.01), non-fasting glucose (p < 0.05 to p < 0.001) and triacylglycerol deposition in liver and adipose tissue (p < 0.001). All treatment regimens improved glucose tolerance (p < 0.05 to p < 0.001) and insulin sensitivity (p < 0.001). Combined treatment with (pGlu-Gln)-CCK-8 and [D-Leu-4]-OB3 resulted in significantly lowered plasma insulin levels, normalisation of circulating LDL-cholesterol and decreased triacylglycerol deposition in muscle. These effects were superior to either treatment regimen alone. There were no changes in overall locomotor activity or respiratory exchange ratio, but treatment with (pGlu-Gln)-CCK-8 significantly reduced (p < 0.001) energy expenditure. CONCLUSIONS/INTERPRETATION: These studies highlight the potential of (pGlu-Gln)-CCK-8 alone and in combination with [D-Leu-4]-OB3 in the treatment of obesity and diabetes.


Subject(s)
Leptin/metabolism , Receptor, Cholecystokinin A/metabolism , Receptors, Cholecystokinin/metabolism , Animals , Area Under Curve , Blood Glucose/metabolism , Body Weight , Calorimetry, Indirect , Chemokines, CC , Diet, High-Fat , Insulin/metabolism , Insulin Resistance , Mice , Obesity/complications , Obesity/metabolism , Peptides/chemistry , Signal Transduction , Time Factors , Up-Regulation
19.
Am J Physiol Gastrointest Liver Physiol ; 304(8): G749-61, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23392236

ABSTRACT

Segmentation is an important process in nutrient mixing and absorption; however, the mechanisms underlying this motility pattern are poorly understood. Segmentation can be induced by luminal perfusion of fatty acid in guinea pig small intestine in vitro and mimicked by the serotonin (5-HT) reuptake inhibitor fluoxetine (300 nM) and by cholecystokinin (CCK). Serotonergic and CCK-related mechanisms underlying nutrient-induced segmentation were investigated using selective 5-HT and CCK receptor antagonists on isolated segments of small intestine luminally perfused with 1 mM decanoic acid. Motility patterns were analyzed using video imaging and spatiotemporal maps. Segmenting activity mediated by decanoic acid was depressed following luminal application of the 5-HT receptor antagonists granisetron (5-HT(3), 1 µM) and SB-207266 (5-HT(4), 10 nM) and the CCK receptor antagonists devazepide (CCK-1, 300 nM) and L-365260 (CCK-2, 300 nM), but these antagonists did not further depress segmentation when combined. The P2 receptor antagonist pyridoxal phosphate-6-azophenyl-2',4'-disulfonate (10 µM) had no effect on activity. Serosal application of 5-HT antagonists had little effect on segmentation in the duodenum but reduced activity in the jejunum when granisetron and SB-207266 were applied together. These results reveal that 5-HT(3) and 5-HT(4) receptors, as well as CCK-1 and CCK-2 receptors, are critical in regulating decanoic acid-induced segmentation. Computational simulation indicated that these data are consistent with decanoic acid activating two pathways in the mucosa that converge within the enteric neural circuitry, while contraction-induced release of 5-HT from the mucosa provides feedback into the neural circuit to set the time course of the overall contractile activity.


Subject(s)
Cholecystokinin/metabolism , Gastrointestinal Motility/physiology , Intestine, Small/physiology , Serotonin/metabolism , Animals , Decanoic Acids/metabolism , Enteric Nervous System/drug effects , Enteric Nervous System/physiology , Fatty Acids/metabolism , Female , Fluoxetine/pharmacology , Gastrointestinal Motility/drug effects , Guinea Pigs , Intestine, Small/drug effects , Male , Muscle Contraction/drug effects , Muscle Contraction/physiology , Myoelectric Complex, Migrating/drug effects , Myoelectric Complex, Migrating/physiology , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin B/metabolism , Receptors, Purinergic P2X/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology
20.
Am J Physiol Gastrointest Liver Physiol ; 305(5): G383-91, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23812038

ABSTRACT

Nutritional stimulation of the cholecystokinin-1 receptor (CCK-1R) and nicotinic acetylcholine receptor (nAChR)-mediated vagal reflex was shown to reduce inflammation and preserve intestinal integrity. Mast cells are important early effectors of the innate immune response; therefore modulation of mucosal mast cells is a potential therapeutic target to control the acute inflammatory response in the intestine. The present study investigates intestinal mast cell responsiveness upon nutritional activation of the vagal anti-inflammatory reflex during acute inflammation. Mucosal mast cell degranulation was induced in C57/Bl6 mice by administration of Salmonella enterica LPS. Lipid-rich enteral feeding prior to LPS significantly decreased circulatory levels of mouse mast cell protease at 30 min post-LPS compared with isocaloric low-lipid nutrition or fasting. CCK-1R blockage reversed the inhibitory effects of lipid-rich feeding, whereas stimulation of the peripheral CCK-1R mimicked nutritional mast cell inhibition. The effects of lipid-rich nutrition were negated by nAChR blockers chlorisondamine and α-bungarotoxin and vagal intestinal denervation. Accordingly, release of ß-hexosaminidase by MC/9 mast cells following LPS or IgE-ovalbumin complexes was dose dependently inhibited by acetylcholine and nicotine. Application of GSK1345038A, a specific agonist of the nAChR α7, in bone marrow-derived mast cells from nAChR ß2-/- and wild types indicated that cholinergic inhibition of mast cells is mediated by the nAChR α7 and is independent of the nAChR ß2. Together, the present study reveals mucosal mast cells as a previously unknown target of the nutritional anti-inflammatory vagal reflex.


Subject(s)
Cell Degranulation , Dietary Fats/administration & dosage , Enteral Nutrition , Inflammation/prevention & control , Intestinal Mucosa/immunology , Intestinal Mucosa/innervation , Mast Cells/immunology , Reflex , Vagus Nerve/physiopathology , Animals , Cell Degranulation/drug effects , Cell Line , Cholinergic Agonists/pharmacology , Chymases/blood , Disease Models, Animal , Histamine Antagonists/pharmacology , Immunity, Mucosal , Inflammation/blood , Inflammation/immunology , Inflammation/physiopathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipopolysaccharides , Male , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nicotinic Antagonists/pharmacology , Receptor, Cholecystokinin A/metabolism , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Vagotomy, Proximal Gastric , Vagus Nerve/drug effects , Vagus Nerve/immunology , Vagus Nerve/metabolism , Vagus Nerve/surgery , beta-N-Acetylhexosaminidases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL