Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Am J Physiol Regul Integr Comp Physiol ; 313(5): R535-R548, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28768657

ABSTRACT

Apolipoprotein AIV (ApoAIV) and cholecystokinin (CCK) are well-known satiating signals that are stimulated by fat consumption. Peripheral ApoAIV and CCK interact to prolong satiating signals. In the present study, we hypothesized that ApoAIV and CCK control energy homeostasis in response to high-fat diet feeding. To test this hypothesis, energy homeostasis in ApoAIV and CCK double knockout (ApoAIV/CCK-KO), ApoAIV knockout (ApoAIV-KO), and CCK knockout (CCK-KO) mice were monitored. When animals were maintained on a low-fat diet, ApoAIV/CCK-KO, ApoAIV-KO, and CCK-KO mice had comparable energy intake and expenditure, body weight, fat mass, fat absorption, and plasma parameters relative to the controls. In contrast, these KO mice exhibited impaired lipid transport to epididymal fat pads in response to intraduodenal infusion of dietary lipids. Furthermore, ApoAIV-KO mice had upregulated levels of CCK receptor 2 (CCK2R) in the small intestine while ApoAIV/CCK-KO mice had upregulated levels of CCK2R in the brown adipose tissue. After 20 wk of a high-fat diet, ApoAIV-KO and CCK-KO mice had comparable body weight and fat mass, as well as lower energy expenditure at some time points. However, ApoAIV/CCK-KO mice exhibited reduced body weight and adiposity relative to wild-type mice, despite having normal food intake. Furthermore, ApoAIV/CCK-KO mice displayed normal fat absorption and locomotor activity, as well as enhanced energy expenditure. These observations suggest that mice lacking ApoAIV and CCK have reduced body weight and adiposity, possibly due to impaired lipid transport and elevated energy expenditure.


Subject(s)
Apolipoproteins A/metabolism , Cholecystokinin/metabolism , Dietary Fats/metabolism , Homeostasis/physiology , Adiposity/genetics , Adiposity/physiology , Animals , Apolipoproteins A/deficiency , Body Weight/physiology , Cholecystokinin/deficiency , Diet, Fat-Restricted/methods , Eating/physiology , Energy Intake/physiology , Energy Metabolism/genetics , Mice, Knockout
2.
Neuron ; 89(6): 1194-1207, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26948890

ABSTRACT

SCA1, a fatal neurodegenerative disorder, is caused by a CAG expansion encoding a polyglutamine stretch in the protein ATXN1. We used RNA sequencing to profile cerebellar gene expression in Pcp2-ATXN1[82Q] mice with ataxia and progressive pathology and Pcp2-ATXN1[30Q]D776 animals having ataxia in absence of Purkinje cell progressive pathology. Weighted Gene Coexpression Network Analysis of the cerebellar expression data revealed two gene networks that significantly correlated with disease and have an expression profile correlating with disease progression in ATXN1[82Q] Purkinje cells. The Magenta Module provides a signature of suppressed transcriptional programs reflecting disease progression in Purkinje cells, while the Lt Yellow Module reflects transcriptional programs activated in response to disease in Purkinje cells as well as other cerebellar cell types. Furthermore, we found that upregulation of cholecystokinin (Cck) and subsequent interaction with the Cck1 receptor likely underlies the lack of progressive Purkinje cell pathology in Pcp2-ATXN1[30Q]D776 mice.


Subject(s)
Ataxin-1/genetics , Cerebellum/metabolism , Cerebellum/pathology , Spinocerebellar Ataxias/pathology , Transcriptome/genetics , Animals , Ataxin-1/metabolism , Chemokines, CC/deficiency , Chemokines, CC/genetics , Cholecystokinin/deficiency , Cholecystokinin/genetics , Disease Models, Animal , Disease Progression , Gene Regulatory Networks , Guanine Nucleotide Exchange Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuropeptides/metabolism , Nuclear Proteins/metabolism , Peptides/genetics , Peptides/metabolism , Purkinje Cells/metabolism , Receptor, Cholecystokinin B/deficiency , Receptor, Cholecystokinin B/genetics , Up-Regulation/genetics
3.
Neurogastroenterol Motil ; 28(3): 364-75, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26604077

ABSTRACT

BACKGROUND: Celiac disease is an autoimmune enteropathy caused by a permanent intolerance to dietary gluten in genetically predisposed individuals. Cholecystokinin (CCK) release from the proximal small intestine and gallbladder emptying in response to a fatty meal are greatly reduced in celiac patients before they start the gluten-free diet, showing a genetic predisposition to gallstones. METHODS: To elucidate the complex pathophysiological mechanisms determining the biliary characteristic of celiac disease, we investigated the effect of the absence of endogenous CCK on cholesterol crystallization and gallstone formation in mice fed a lithogenic diet for 28 days. KEY RESULTS: Fasting gallbladder volumes were increased and the response of gallbladder emptying to the high-fat diet was impaired in CCK knockout (KO) mice compared to wild-type mice. Because of the absence of CCK, small intestinal transit time was prolonged and intestinal cholesterol absorption was increased. During 28 days of feeding, elevated biliary cholesterol concentrations and gallbladder stasis promoted the growth and agglomeration of solid cholesterol crystals into microlithiasis and stones. Thus, cholesterol crystallization and gallstone formation were accelerated in CCK KO mice. In contrast, daily intraperitoneal administration of CCK-8 reduced gallstone formation in CCK KO mice even on the lithogenic diet. CONCLUSIONS & INFERENCES: The lack of endogenous CCK enhances susceptibility to gallstones by impairing gallbladder contractile function and small intestinal motility function. These findings show that celiac disease is an important risk factor for gallstone formation and the gallbladder motility function should be routinely examined by ultrasonography and gallbladder stasis should be prevented in celiac patients.


Subject(s)
Cholecystokinin/deficiency , Gallbladder Emptying/physiology , Gallstones/etiology , Animals , Bile/chemistry , Bile/metabolism , Celiac Disease/complications , Gallstones/metabolism , Gastrointestinal Transit/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction
4.
Neuroscience ; 218: 185-95, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22613736

ABSTRACT

Cholecystokinin (CCK) is a neuropeptide widely distributed in the mammalian brain. This peptide regulates many physiological functions and behaviors, such as cardio-respiratory control, thermoregulation, nociception, feeding, memory processes and motivational responses, and plays a prominent role in emotional responses including anxiety and depression. CCK-expressing brain regions involved in these functions remain unclear and their identification represents an important step towards understanding CCK function in the brain. The basolateral amygdala (BLA) is strongly involved in emotional processing and expresses high levels of CCK. In this study we examined the contribution of CCK expressed in this brain region to emotional responses in mice. To knockdown CCK specifically in the BLA, we used stereotaxic delivery of recombinant adeno-associated viral vectors expressing a CCK-targeted shRNA. This procedure efficiently reduced CCK levels locally. shCCK-treated animals showed reduced levels of anxiety in the elevated plus-maze, and lower despair-like behavior in the forced swim test. Our data demonstrate that CCK expressed in the BLA represents a key brain substrate for anxiogenic and depressant effects of the peptide. The study also suggests that elevated amygdalar CCK could contribute to panic and major depressive disorders that have been associated with CCK dysfunction in humans.


Subject(s)
Amygdala/metabolism , Anxiety/metabolism , Cholecystokinin/metabolism , Depression/metabolism , Animals , Cholecystokinin/deficiency , Gene Knockdown Techniques , Image Processing, Computer-Assisted , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
5.
J Neurophysiol ; 106(3): 1515-24, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21753024

ABSTRACT

Cholecystokinin (CCK) is one of the most abundant neuropeptides in the brain, where it interacts with two G protein-coupled receptors (CCK-1 and CCK-2). Activation of both CCK receptors increases the activity of PLC, resulting in increases in intracellular calcium ion (Ca(2+)) release and activation of PKC. Whereas high density of CCK receptors has been detected in the superficial layers of the entorhinal cortex (EC), the functions of CCK in this brain region have not been determined. Here, we studied the effects of CCK on neuronal excitability of layer III pyramidal neurons in the EC. Our results showed that CCK remarkably increased the firing frequency of action potentials (APs). The effects of CCK on neuronal excitability were mediated via activation of CCK-2 receptors and required the functions of G proteins and PLC. However, CCK-mediated facilitation of neuronal excitability was independent of inositol trisphosphate receptors and PKC. CCK facilitated neuronal excitability by activating a cationic channel to generate membrane depolarization. The effects of CCK were suppressed by the generic, nonselective cationic channel blockers, 2-aminoethyldiphenyl borate and flufenamic acid, but potentiated by gadolinium ion and lanthanum ion at 100 µM. Depletion of extracellular Ca(2+) also counteracted CCK-induced increases in AC firing frequency. Moreover, CCK-induced enhancement of neuronal excitability was inhibited significantly by intracellular application of the antibody to transient receptor potential channel 5 (TRPC5), suggesting the involvement of TRPC5 channels. Our results provide a cellular and molecular mechanism to help explain the functions of CCK in vivo.


Subject(s)
Cholecystokinin/physiology , Entorhinal Cortex/physiology , Neurons/physiology , TRPC Cation Channels/physiology , Animals , Antibodies/toxicity , Cholecystokinin/antagonists & inhibitors , Cholecystokinin/deficiency , Mice , Mice, Knockout , Neurons/immunology , Pyramidal Cells/immunology , Pyramidal Cells/physiology , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin B/deficiency , Receptor, Cholecystokinin B/genetics , TRPC Cation Channels/immunology
6.
Diabetes ; 60(7): 2000-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21602512

ABSTRACT

OBJECTIVE: Cholecystokinin (CCK) is released in response to lipid intake and stimulates insulin secretion. We hypothesized that CCK deficiency would alter the regulation of insulin secretion and glucose homeostasis. RESEARCH DESIGN AND METHODS: We used quantitative magnetic resonance imaging to determine body composition and studied plasma glucose and insulin secretion of CCK gene knockout (CCK-KO) mice and their wild-type controls using intraperitoneal glucose and arginine infusions. The area of anti-insulin staining in pancreatic islets was measured by immunohistochemistry. Insulin sensitivity was assessed with euglycemic-hyperinsulemic clamps. RESULTS: CCK-KO mice fed a low-fat diet had a reduced acute insulin response to glucose but a normal response to arginine and normal glucose tolerance, associated with a trend toward greater insulin sensitivity. However, when fed a high-fat diet (HFD) for 10 weeks, CCK-KO mice developed glucose intolerance despite increased insulin sensitivity that was associated with low insulin secretion in response to both glucose and arginine. The deficiency of insulin secretion in CCK-KO mice was not associated with changes in ß-cell or islet size. CONCLUSIONS: CCK is involved in regulating insulin secretion and glucose tolerance in mice eating an HFD. The impaired insulin response to intraperitoneal stimuli that do not typically elicit CCK release suggests that this hormone has chronic effects on ß-cell adaptation to diet in addition to acute incretin actions.


Subject(s)
Cholecystokinin/deficiency , Insulin/metabolism , Insulin/physiology , Animals , Arginine , Diet, Fat-Restricted , Dietary Fats/administration & dosage , Glucose/metabolism , Glucose Intolerance/chemically induced , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/drug effects , Mice , Mice, Knockout
7.
Eur J Neurosci ; 32(6): 1006-17, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20731710

ABSTRACT

The suprachiasmatic nucleus (SCN) is the principal pacemaker driving circadian rhythms of physiology and behaviour. Neurons within the SCN express both classical and neuropeptide transmitters which regulate clock functions. Cholecyctokinin (CCK) is a potent neurotransmitter expressed in neurons of the mammalian SCN, but its role in circadian timing is not known. In the present study, CCK was demonstrated in a distinct population of neurons located in the shell region of the SCN and in a few cells in the core region. The CCK neurons did not express vasopressin or vasoactive intestinal peptide. However, CCK-containing processes make synaptic contacts with both groups of neurons and some CCK cell bodies were innervated by VIPergic neurons. The CCK neurons received no direct input from the three major pathways to the SCN, and the CCK neurons were not light-responsive as evaluated by induction of cFOS, and did not express the core clock protein PER1. Accordingly, CCK-deficient mice showed normal entrainment and had similar τ, light-induced phase shift and negative masking behaviour as wild-type animals. In conclusion, CCK signalling seems not to be involved directly in light-induced resetting of the clock or in regulating core clock function. The expression of CCK in a subpopulation of neurons, which do not belonging to either the VIP or AVP cells but which have synaptic contacts to both cell types and reverse innervation of CCK neurons from VIP neurons, suggests that the CCK neurons may act in non-photic regulation within the clock and/or, via CCK projections, mediate clock information to hypothalamic nuclei.


Subject(s)
Cholecystokinin/deficiency , Cholecystokinin/physiology , Circadian Rhythm/physiology , Gene Expression Regulation/physiology , Lighting , Neurons/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Cholecystokinin/biosynthesis , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/chemistry , Suprachiasmatic Nucleus/chemistry
8.
Gastroenterology ; 138(5): 1997-2005, 2010 May.
Article in English | MEDLINE | ID: mdl-20117110

ABSTRACT

BACKGROUND & AIMS: Cholecystokinin (CCK) is a satiation peptide released during meals in response to lipid intake; it regulates pancreatic digestive enzymes that are required for absorption of nutrients. We proposed that mice with a disruption in the CCK gene (CCK knockout [CCK-KO] mice) that were fed a diet of 20% butter fat would have altered fat metabolism. METHODS: We used quantitative magnetic resonance imaging to determine body composition and monitored food intake of CCK-KO mice using an automated measurement system. Intestinal fat absorption and energy expenditure were determined using a noninvasive assessment of intestinal fat absorption and an open circuit calorimeter, respectively. RESULTS: After consuming a high-fat diet for 10 weeks, CCK-KO mice had reduced body weight gain and body fat mass and enlarged adipocytes, despite the same level of food intake as wild-type mice. CCK-KO mice also had defects in fat absorption, especially of long-chain saturated fatty acids, but pancreatic triglyceride lipase did not appear to have a role in the fat malabsorption. Energy expenditure was higher in CCK-KO than wild-type mice, and CCK-KO mice had greater oxidation of carbohydrates while on the high-fat diet. Plasma leptin levels in the CCK-KO mice fed the high-fat diet were markedly lower than in wild-type mice, although levels of insulin, gastric-inhibitory polypeptide, and glucagon-like peptide-1 were normal. CONCLUSIONS: CCK is involved in regulating the metabolic rate and is important for lipid absorption and control of body weight in mice placed on a high-fat diet.


Subject(s)
Cholecystokinin/deficiency , Dietary Fats/metabolism , Intestinal Absorption , Obesity/prevention & control , Weight Gain , Adiposity , Animals , Biomarkers/blood , Butter , Calorimetry , Cholecystokinin/genetics , Dietary Fats/blood , Disease Models, Animal , Eating , Energy Metabolism , Fatty Acids/metabolism , Leptin/blood , Lipase/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology , Time Factors
9.
Curr Top Med Chem ; 7(12): 1211-31, 2007.
Article in English | MEDLINE | ID: mdl-17584143

ABSTRACT

In this paper, the established and possible roles of CCK1 and CCK2 receptors in gastrointestinal (GI) and metabolic diseases are reviewed and available results from human agonist/antagonist studies are discussed. While there is evidence for the involvement of CCK1R in numerous diseases including pancreatic disorders, motility disorders, tumor growth, regulation of satiety and a number of CCK-deficient states, the role of CCK1R in these conditions is not clearly defined. There are encouraging data from several clinical studies of CCK1R antagonists in some of these conditions, but their role as therapeutic agents remains unclear. The role of CCK2R in physiological (atrophic gastritis, pernicious anemia) and pathological (Zollinger-Ellison syndrome) hypergastrinemic states, its effects on the gastric mucosa (ECL cell hyperplasia, carcinoids, parietal cell mass) and its role in acid-peptic disorders are clearly defined. Furthermore, recent studies point to a possible role for CCK2R in a number of GI malignancies. Current data from human studies of CCK2R antagonists are presented and their potential role in the treatment of these conditions reviewed. Furthermore, the role of CCK2 receptors as targets for medical imaging is discussed.


Subject(s)
Gastrins/metabolism , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/metabolism , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Receptors, Cholecystokinin/metabolism , Animals , Cholecystokinin/deficiency , Cholecystokinin/genetics , Cholecystokinin/metabolism , Gastrointestinal Diseases/pathology , Humans , Metabolic Diseases/pathology , Receptors, Cholecystokinin/agonists , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/genetics
10.
Am J Physiol Gastrointest Liver Physiol ; 292(2): G667-77, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17095753

ABSTRACT

Endogenous CCK release induced by a synthetic trypsin inhibitor, camostat, stimulates pancreatic growth; however, the mechanisms mediating this growth are not well established. Early response genes often couple short-term signals with long-term responses. To study their participation in the pancreatic growth response, mice were fasted for 18 h and refed chow containing 0.1% camostat for 1-24 h. Expression of 18 early response genes were evaluated by quantitative PCR; mRNA for 17 of the 18 increased at 1, 2, 4, or 8 h. Protein expression for c-jun, c-fos, ATF-3, Egr-1, and JunB peaked at 2 h. Nuclear localization was confirmed by immunohistochemistry of c-fos, c-jun, and Egr-1. Refeeding regular chow induced only a small increase of c-jun and none in c-fos expression. JNKs and ERKs were activated 1 h after camostat feeding as was the phosphorylation of c-jun and ATF-2. AP-1 DNA binding evaluated by EMSA showed a significant increase 1-2 h after camostat feeding with participation of c-jun, c-fos, ATF-2, ATF-3, and JunB shown by supershift. The CCK antagonist IQM-95,333 blocked camostat feeding-induced c-jun and c-fos expression by 67 and 84%, respectively, and AP-1 DNA binding was also inhibited. In CCK-deficient mice, the maximal response of c-jun induction and AP-1 DNA binding were reduced by 64 and 70%, respectively. These results indicate that camostat feeding induces a spectrum of early response gene expression and AP-1 DNA binding and that these effects are mainly CCK dependent.


Subject(s)
Gene Expression/drug effects , Immediate-Early Proteins/genetics , Pancreas/drug effects , Trypsin Inhibitors/pharmacology , Activating Transcription Factor 2/metabolism , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Carbamates/pharmacology , Cholecystokinin/deficiency , Cholecystokinin/genetics , Early Growth Response Transcription Factors/genetics , Early Growth Response Transcription Factors/metabolism , Esters , Extracellular Signal-Regulated MAP Kinases/metabolism , Gabexate/analogs & derivatives , Gabexate/pharmacology , Guanidines , Immediate-Early Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Mice, Inbred ICR , Mice, Knockout , Pancreas/growth & development , Pancreas/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Pyrimidinones/pharmacology , Receptors, Cholecystokinin/antagonists & inhibitors , Transcription Factor AP-1/metabolism
11.
J Nutr ; 136(7): 1792-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16772439

ABSTRACT

Feeding stimulates pancreatic digestive enzyme synthesis at the translational level, and this is thought to be mediated by hormones and neurotransmitters. However, BCAAs, particularly leucine, stimulate protein synthesis in several tissues. We investigated whether BCAA stimulated the translational machinery in murine pancreas and whether their effects were independent of hormones. Rats and mice were administered (i.g. gavage) individual BCAA at 1.35 mg/g (body weight) and rat isolated pancreatic acini were incubated with BCAA under different conditions. Activation of translation initiation factors and total protein synthesis were analyzed. BCAA gavage stimulated the phosphorylation of the initiation factor 4E (eIF4E) binding protein 1 (4E-BP1) and the ribosomal protein S6 kinase (S6K), with leucine being the most effective. Leucine also increased the association of the initiation factors eIF4E and eIF4G, but did not affect the activity of the guanine nucleotide exchange factor eIF2B, nor total protein synthesis. BCAA acted independently of insulin signaling on isolated pancreatic acini from diabetic rats. The ability of leucine to promote phosphorylation of 4E-BP1 and S6K as well as enhance the assembly of the eIF4F complex was unimpaired in CCK-deficient mice. Finally, rapamycin (0.75 mg/kg) administered to rats 2 h before leucine gavage inhibited the phosphorylation of S6 and 4E-BP1 induced by leucine. We conclude that leucine may participate, as a signal as well as a substrate, in activating the translational machinery in pancreatic acinar cells independently of hormonal effects and that this action is through the mTOR pathway.


Subject(s)
Amino Acids, Branched-Chain/pharmacology , DNA-Binding Proteins/biosynthesis , Leucine/pharmacology , Pancreas, Exocrine/drug effects , Protein Kinases/drug effects , Ribosomal Protein S6 Kinases/biosynthesis , Transcription Factors/biosynthesis , Amino Acids, Branched-Chain/blood , Amino Acids, Branched-Chain/metabolism , Animals , Cholecystokinin/deficiency , Cholecystokinin/physiology , DNA-Binding Proteins/metabolism , Drug Interactions , Immunosuppressive Agents/pharmacology , Insulin/blood , Leucine/metabolism , Leucine/physiology , Male , Mice , Mice, Inbred ICR , Pancreas, Exocrine/metabolism , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transcription Factors/metabolism
12.
Endocrinology ; 146(10): 4464-71, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16002530

ABSTRACT

The antral hormone gastrin and its intestinal relative, cholecystokinin (CCK), are pivotal in the regulation of gastric functions. Other gastric hormones like ghrelin, peptide YY (PYY), and islet amyloid polypeptide (IAPP), however, also contribute to the regulation of acid secretion, motility, and feeding. Because gastrin and CCK are crucial for gastric homeostasis, we examined how loss of gastrin alone and gastrin plus CCK affected the expression of ghrelin, IAPP, and PYY and ghrelin secretion. The expression of ghrelin, IAPP, and PYY and the CCK-A receptor genes were examined in both gastrin and gastrin-CCK double-knockout (KO) mice using immunocytochemistry and quantitative RT-PCR. Ghrelin concentrations in plasma were measured using RIA. Gastrin and CCK were infused in gastrin-CCK KO mice using osmotic minipumps. The number of ghrelin cells and ghrelin gene expression were unaffected, albeit the ghrelin cells were located closer to the base of the glands in both KO mouse strains when freely fed. However, lack of both gastrin and CCK attenuated fasting-induced ghrelin expression and secretion. Fundic ghrelin cells expressed the CCK-A receptor, and ghrelin expression increased after CCK infusion. Furthermore, gastric IAPP and PYY expression as well as the number of IAPP- and PYY-containing cells were reduced in both gastrin and gastrin-CCK KO mice. Gastrin infusion increased gastric IAPP but not PYY expression. In conclusion, lack of gastrin plus CCK but not gastrin alone reduced ghrelin secretion in response to fasting through both direct and indirect mechanisms. Both gastrin and combined gastrin-CCK deficiency reduced the gastric IAPP and PYY expression.


Subject(s)
Amyloid/genetics , Cholecystokinin/deficiency , Gastrins/deficiency , Gene Expression Regulation , Peptide Hormones/genetics , Peptide YY/genetics , Amyloid/blood , Animals , Cholecystokinin/administration & dosage , Cholecystokinin/genetics , Cholecystokinin/pharmacology , Fasting , Gastric Fundus/physiology , Gastrins/administration & dosage , Gastrins/genetics , Gastrins/pharmacology , Ghrelin , Infusions, Parenteral , Islet Amyloid Polypeptide , Mice , Mice, Knockout , Peptide Hormones/blood , Polymerase Chain Reaction , Pyloric Antrum/physiology , RNA, Messenger/drug effects , RNA, Messenger/genetics
13.
Gastroenterology ; 126(2): 476-87, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14762785

ABSTRACT

BACKGROUND & AIMS: Three pathways control gastric acid secretion: the gastrin-enterochromaffin-like (ECL) cell axis, the vagus-parietal cell axis, and the cholecystokinin (CCK)-D cell axis. Mice lacking gastrin or both gastrin and CCK were examined to determine the role of the hormones. METHODS: Acid was measured after pylorus ligation, and biopsies from gastrin knockout (KO), gastrin-CCK double-KO, and wild-type (WT) mice were collected for biochemical, immunocytochemical, and electron-microscopic examination. RESULTS: The ECL cells were inactive in both groups of mutant mice but the cell number was unaffected. Both parietal cell number and level of H(+)/K(+)-ATPase messenger RNA (mRNA) were reduced in the mutant strains, but gastrin-CCK double-KO mice displayed more active parietal cells and larger acid output than the gastrin KO mice. The acid response to histamine in double-KO mice was unchanged whereas that to gastrin was diminished, but it could be restored by infusion of gastrin. Oxyntic D-cell density was the same in both mutant strains, but the D cells were more active in the gastrin KO than in the double-KO mice. CCK infusion in gastrin-CCK double-KO mice raised the somatostatin mRNA level and inhibited acid secretion to the level seen in gastrin KO mice. Vagotomy and atropine abolished acid secretion in all 3 groups of mice. CONCLUSIONS: Lack of gastrin impairs the gastrin-ECL axis, whereas lack of gastrin and CCK impairs both hormonal pathways. In the gastrin-CCK double-KO mice, acid secretion is only controlled by cholinergic vagal stimulation, which normalizes the acid output.


Subject(s)
Cholecystokinin/deficiency , Gastric Acid/metabolism , Gastrins/deficiency , Animals , Enterochromaffin Cells/metabolism , Mice , Mice, Knockout , Parietal Cells, Gastric/metabolism , Somatostatin-Secreting Cells/metabolism , Vagotomy
14.
Neuroscience ; 118(1): 19-23, 2003.
Article in English | MEDLINE | ID: mdl-12676133

ABSTRACT

The central fragment of cholecystokinin, CCK8, plays a critical role in stress-related changes in behavior and memory. Therefore, we investigated whether the endogenous cholecystokininergic system is involved in the impairment of attention and/or memory induced by stressful conditions. Plasma corticosterone concentrations increased three-fold and plasma adrenocorticotropin (ACTH); concentrations increased five-fold when rats were maintained in the open arm of an elevated plus maze for 5 min. The same stress conditions impaired spatial recognition in the two-trial memory task. In addition, this stress led to a significant decrease in the extracellular levels of cholecystokinin-like immunoreactivity in the dorsal subiculum/CA1 of the hippocampus and partially suppressed the increase obtained during the acquisition phase of memory. This suggests that the cholecystokininergic system in the hippocampus is involved in stress-induced impairment of spatial recognition memory.


Subject(s)
Brain/metabolism , Cholecystokinin/deficiency , Memory Disorders/metabolism , Neural Pathways/metabolism , Stress, Physiological/metabolism , Adrenocorticotropic Hormone/blood , Animals , Brain/physiopathology , Corticosterone/blood , Down-Regulation/physiology , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Maze Learning/physiology , Memory Disorders/physiopathology , Neural Pathways/physiopathology , Rats , Rats, Wistar , Stress, Physiological/physiopathology
15.
Pharmacol Toxicol ; 91(6): 333-50, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12688377

ABSTRACT

In this paper the possible roles of cholecystokinin (CCK), gastrin, or gastrin-related peptides and their receptors in human gastrointestinal diseases are reviewed. For CCK/CCK(A) receptors (CCK(A)-R), the evidence for their proposed involvement in diseases caused by impaired CCK release or CCK(A)-R mutations, pancreatic disorders (acute/chronic pancreatitis), gastrointestinal motility disorders (gallbladder disease, irritable bowel syndrome), pancreatic tumor growth and satiety disorders, is briefly reviewed. The evidence that has established the involvement of gastrin/CCK(B)-R in mediating the action of hypergastrinaemic disorders, mediating hypergastrinaemic effects on the gastric mucosa (ECL hyperplasia, carcinoids, parietal cell mass), and acid-peptic diseases, is reviewed. The evidence for their possible involvement in mediating growth of gastric and pancreatic tumours and possible involvement of gastrin-related peptides in colon cancers, is reviewed briefly.


Subject(s)
Cholecystokinin , Gastrins , Gastrointestinal Diseases/metabolism , Gastrointestinal Neoplasms/etiology , Pancreatic Neoplasms/etiology , Receptors, Cholecystokinin , Animals , Cholecystokinin/deficiency , Cholecystokinin/metabolism , Cholecystokinin/physiology , Gastrins/metabolism , Gastrins/physiology , Gastrointestinal Diseases/etiology , Gastrointestinal Motility/physiology , Gastrointestinal Neoplasms/metabolism , Humans , Pancreatic Neoplasms/metabolism , Receptors, Cholecystokinin/deficiency , Receptors, Cholecystokinin/metabolism , Receptors, Cholecystokinin/physiology
16.
Article in English | MEDLINE | ID: mdl-11713979

ABSTRACT

The gastric hormone gastrin was first recognized for its ability to induce acid secretion. Following the purification and subsequent development of specific radioimmunoassays for gastrin, it was also shown to be a regulator of oxyntic mucosal growth. To examine the importance of gastrin or its receptors during development in general and for gastric physiology specifically both have been knocked out. Gastrin and gastrin receptor knockout mice are viable, develop without any gross abnormalities, and are fertile. Even though gastrin acts as a growth factor during hypergastrinemia there was no general atrophy of the gastric mucosa in the knockout mice. However, the maturation of both parietal and ECL cells was disturbed and the number of parietal cells was reduced. Basal acid secretion was impaired and rendered the parietal cells unresponsive to secretagogues. Outside the stomach the mice had no apparent phenotype. However, studies have suggested that progastrin and glycine-extended proforms of gastrin may have biological importance, but these results are still circumstantial and identification of the implicated receptors will be crucial for further studies.


Subject(s)
Gastrins/deficiency , Gastrins/genetics , Receptors, Cholecystokinin/deficiency , Receptors, Cholecystokinin/genetics , Animals , Cholecystokinin/deficiency , Cholecystokinin/genetics , Cholecystokinin/physiology , Colorectal Neoplasms/etiology , Gastric Mucosa/growth & development , Gastric Mucosa/physiology , Gastrins/physiology , Gene Expression , Mice , Mice, Knockout , Phenotype , Receptors, Cholecystokinin/physiology
20.
Am J Physiol ; 276(5): G1302-9, 1999 05.
Article in English | MEDLINE | ID: mdl-10330022

ABSTRACT

A CCK-deficient mouse mutant generated by gene targeting in embryonic stem cells was analyzed to determine the importance of CCK for growth and function of the exocrine pancreas and for pancreatic adaptation to dietary changes. RIAs confirmed the absence of CCK in mutant mice and demonstrated that tissue concentrations of the related peptide gastrin were normal. CCK-deficient mice are viable and fertile and exhibit normal body weight. Pancreas weight and cellular morphology appeared normal, although pancreatic amylase content was elevated in CCK-deficient mice. We found that a high-protein diet increased pancreatic weight, protein, DNA, and chymotrypsinogen content similarly in CCK-deficient and wild-type mice. This result demonstrates that CCK is not required for protein-induced pancreatic hypertrophy and increased proteolytic enzyme content. This is a novel finding, since CCK has been considered the primary mediator of dietary protein-induced changes in the pancreas. Altered somatostatin concentrations in brain and duodenum of CCK-deficient mice suggest that other regulatory pathways are modified to compensate for the CCK deficiency.


Subject(s)
Cholecystokinin/deficiency , Cholecystokinin/physiology , Dietary Proteins/administration & dosage , Pancreas/physiology , Adaptation, Physiological , Amylases/analysis , Animals , Cholecystokinin/genetics , Chymotrypsinogen/analysis , Digestive System/chemistry , Female , Gastrins/analysis , Gene Targeting , Male , Mice , Mutagenesis , Organ Size , Pancreas/cytology , Pancreas/enzymology , RNA, Messenger/analysis , Receptor, Cholecystokinin A , Receptors, Cholecystokinin/genetics , Somatostatin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...