Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 252
1.
Clin Chem ; 68(4): 584-594, 2022 03 31.
Article En | MEDLINE | ID: mdl-35015868

BACKGROUND: Peptide Tyr-Tyr (PYY1-36), pancreatic polypeptide (PP1-36) and neuropeptide Y (NPY1-36) constitute the PP-fold family of peptides that is involved in metabolic regulation. Very low plasma concentrations and cleavage into active 3-36 fragments challenge bioanalytical assays used for the quantification of these peptides. METHODS: We developed a multiplexed isotopic dilution assay to quantify PYY1-36, PP1-36, and NPY1-36 and their dipeptidyl peptidase-4 (DPP4)-derived metabolites PYY3-36, PP3-36 and NPY3-36. All peptides were immunocaptured from plasma using a monoclonal antibody and quantified by micro-ultra-HPLC-MS/MS. Blood samples from healthy volunteers were collected fasting and 30 min after nutrient stimulation. Method comparison was performed with commercial immunoassays. RESULTS: Linearity was shown in the measured intervals (r2 > 0.99). The lower limit of quantification (LLOQ) with a CV at 20% was 1.5 pM for PYY1-36 and PYY3-36, 3.0 pM for PP1-36 and PP3-36, 0.8 pM for NPY1-36 and 0.5 pM for NPY3-36. In all cases, intra- and inter-assay bias and imprecision were <21%. Pre-analytical stability required addition of a protease inhibitor cocktail. Physiological concentrations of PYY3-36, NPY3-36, PP1-36 and PP3-36 were above the LLOQ in 43% to 100% of the samples. PYY1-36 and NPY1-36 were above the LLOQ in 9% and 0% of the samples, respectively. Immunoassays showed higher concentrations of measurands and poor agreement when compared with micro-UHPLC-MS/MS. CONCLUSIONS: The assay allowed for specific multiplexed analysis of the PP-fold family of peptides and their DPP4-cleaved fragments in a single sample, thereby offering new perspectives to study the role and therapeutic potential of these essential peptide hormones in health and metabolic disease.


Pancreatic Polypeptide , Tandem Mass Spectrometry , Chromatography, High Pressure Liquid , Chromatography, Liquid , Humans , Neuropeptide Y , Pancreatic Polypeptide/pharmacology
2.
Article En | MEDLINE | ID: mdl-27381051

BACKGROUND: Pancreatic polypeptide (PP) is supposed to be one of the major endogenous agonists of the neuropeptide Y4 receptor. Pancreatic polypeptide can influence gastrointestinal motility, acting mainly through vagal mechanisms, but whether PP acts directly on the stomach has not been explored yet. The aims of this study were to investigate the effects of PP on mouse gastric emptying, on spontaneous tone of whole stomach in vitro and to examine the mechanism of action. METHODS: Gastric emptying was measured by red phenol method after i.p. PP administration (1-3 nmol per mouse). Responses induced by PP (1-300 mmol L-1 ) on gastric endoluminal pressure were analyzed in vitro in the presence of different drugs. Gastric genic expression of Y4 receptor was verified by RT-PCR. KEY RESULTS: Pancreatic polypeptide dose-dependently increased non-nutrient liquid gastric emptying rate. In vitro, PP produced a concentration-dependent contraction that was abolished by tetrodotoxin, a neural blocker of Na+ voltage-dependent channels. The contractile response was significantly reduced by atropine, a muscarinic receptor antagonist, and by SR48968, an NK2 receptor antagonist, while it was potentiated by neostigmine, an inhibitor of acetylcholinesterase. The joint application of atropine and SR48968 fully abolished PP contractile effect. Reverse transcriptase-polymerase chain reaction analysis revealed the presence of Y4 receptor mRNA in mouse stomach with a greater expression in antrum than in fundus. CONCLUSIONS & INFERENCES: The present findings demonstrate that exogenous PP stimulates mouse gastric motor activity, by acting directly on the stomach. This effect appears due to the activation of enteric excitatory neurons releasing acetylcholine and tachykinins.


Gastric Emptying/physiology , Gastrointestinal Motility/physiology , Pancreatic Polypeptide/pharmacology , Peripheral Nerves/physiology , Animals , Dose-Response Relationship, Drug , Gastric Emptying/drug effects , Gastrointestinal Motility/drug effects , Male , Mice , Mice, Inbred C57BL , Muscarinic Antagonists/pharmacology , Organ Culture Techniques , Peripheral Nerves/drug effects , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/physiology
3.
Br J Pharmacol ; 173(22): 3208-3221, 2016 11.
Article En | MEDLINE | ID: mdl-27545829

BACKGROUND AND PURPOSE: Human pancreatic polypeptide (hPP) is known to suppress appetite and food intake, thereby representing a potential therapeutic approach against obesity and associated metabolic disorders. The aim of this study was to improve hPP stability by covalent PEGylation with diverse molecular weight polyethylene glycols (PEGs) at two positions using promising lead structures while maintaining target activity. EXPERIMENTAL APPROACH: Modified peptides were synthesized by combined solid-phase and solution-phase peptide synthesis. Their potency was investigated in constitutively expressing human epithelial cells and isolated human colonic mucosa as well as receptor-transfected artificial cell lines. Human blood plasma and porcine liver homogenates were used to examine the in vitro stability of the analogues. The most promising variants were injected s.c. in C57BL/6JRj mice to monitor fasting-induced food intake and bioavailability. KEY RESULTS: In human epithelia and colonic mucosal preparations, activity of the modified hPP peptides depended on the core sequence and latency of the peptides was related to PEG size. Peptides modified with a 22 kDa PEG (PEG22) remained intact in blood plasma and on incubation with liver homogenates for more than 96 h. Finally, hPP2-36 , [K22 (PEG22)]hPP2-36 and [K22 (PEG22),Q34 ]hPP significantly reduced cumulative food intake in mice over 16 h after s.c. administration. CONCLUSIONS AND IMPLICATIONS: Modification with PEG22 at position 22 stabilizes hPP significantly while extending its biological activities and could be used in drug development prospectively.


Eating/drug effects , Pancreatic Polypeptide/metabolism , Pancreatic Polypeptide/pharmacology , Polyethylene Glycols/metabolism , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Molecular Weight , Pancreatic Polypeptide/chemistry
4.
J Med Chem ; 59(13): 6045-58, 2016 07 14.
Article En | MEDLINE | ID: mdl-27223253

The diastereomeric mixture of d/l-2,7-diaminooctanedioyl-bis(YRLRY-NH2) (BVD-74D, 2) was described in the literature as a high affinity Y4 receptor agonist. Here we report on the synthesis and pharmacological characterization of the pure diastereomers (2R,7R)- and (2S,7S)-2 and a series of homo- and heterodimeric analogues in which octanedioic acid was used as an achiral linker. To investigate the role of the Arg residues, one or two arginines were replaced by Ala. Moreover, N(ω)-(6-aminohexylaminocarbonyl)Arg was introduced as an arginine replacement (17). (2R,7R)-2 was superior to (2S,7S)-2 in binding and functional cellular assays and equipotent with 17. [(3)H]Propionylation of one amino group in the linker of (2R,7R)-2 or at the primary amino group in 17 resulted in high affinity Y4R radioligands ([(3)H]-(2R,7R)-10, [(3)H]18) with subnanomolar Kd values.


Pancreatic Polypeptide/chemistry , Pancreatic Polypeptide/pharmacology , Protein Precursors/chemistry , Protein Precursors/pharmacology , Receptors, Neuropeptide Y/agonists , Amino Acid Sequence , Animals , CHO Cells , Cell Line, Tumor , Cricetulus , HEK293 Cells , Humans , Pancreatic Polypeptide/chemical synthesis , Protein Precursors/chemical synthesis , Receptors, Neuropeptide Y/metabolism , Stereoisomerism
5.
Br J Pharmacol ; 173(12): 1925-38, 2016 06.
Article En | MEDLINE | ID: mdl-26844810

BACKGROUND AND PURPOSE: Avoiding danger and finding food are closely related behaviours that are essential for surviving in a natural environment. Growing evidence supports an important role of gut-brain peptides in modulating energy homeostasis and emotional-affective behaviour. For instance, postprandial release of pancreatic polypeptide (PP) reduced food intake and altered stress-induced motor activity and anxiety by activating central Y4 receptors. EXPERIMENTAL APPROACH: We characterized [K(30) (PEG2)]hPP2-36 as long-acting Y4 receptor agonist and injected it peripherally into wildtype and Y4 receptor knockout (Y4KO) C57Bl/6NCrl mice to investigate the role of Y4 receptors in fear conditioning. Extinction and relapse after extinction was measured by spontaneous recovery and renewal. KEY RESULTS: The Y4KO mice showed impaired cued and context fear extinction without affecting acquisition, consolidation or recall of fear. Correspondingly, peripheral injection of [K(30) (PEG2)]hPP2-36 facilitated extinction learning upon fasting, an effect that was long-lasting and generalized. Furthermore, peripherally applied [K(30) (PEG2)]hPP2-36 before extinction inhibited the activation of orexin-expressing neurons in the lateral hypothalamus in WT, but not in Y4KO mice. CONCLUSIONS AND IMPLICATIONS: Our findings suggests suppression of excessive arousal as a possible mechanism for the extinction-promoting effect of central Y4 receptors and provide strong evidence that fear extinction requires integration of vegetative stimuli with cortical and subcortical information, a process crucially depending on Y4 receptors. Importantly, in the lateral hypothalamus two peptide systems, PP and orexin, interact to generate an emotional response adapted to the current homeostatic state. Detailed investigations of feeding-relevant genes may thus deliver multiple intervention points for treating anxiety-related disorders.


Cues , Extinction, Psychological/drug effects , Fear/drug effects , Pancreatic Polypeptide/pharmacology , Receptors, Neuropeptide Y/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Neuropeptide Y/deficiency
6.
Neuropeptides ; 55: 67-71, 2016 Feb.
Article En | MEDLINE | ID: mdl-26601891

Pancreatic polypeptide (PP) is a negative regulator of energy homeostasis that suppresses food intake and lowers body weight. Similar to other gastrointestinal-derived peptides, PP also modulates gastrointestinal motility and may be involved in the regulation of anxiety. Previous studies revealed that PP suppresses gastric emptying but increases colonic motility in mice. In our present study, we assessed the effect of PP on anxiety as well as colonic motility and secretory function. Intracerebroventricular and intravenous routes of PP were administered in conscious rats. Our results showed that intracerebroventricular administration of PP did not affect anxiety in the open field test. Intravenous injection of PP accelerated colonic transit, but did not significantly change fecal amount and fecal fluid composition. On the other hand, intracerebroventricular injection of PP did not alter colonic transit, fecal amount, or fluid composition. In conclusion, peripheral, but not central PP administration enhances colonic motility without eliciting anxiety or altering colonic secretion.


Anxiety/metabolism , Colon/drug effects , Corticotropin-Releasing Hormone/metabolism , Pancreatic Polypeptide/pharmacology , Peptide Fragments/pharmacology , Animals , Colon/metabolism , Gastrointestinal Motility/drug effects , Injections/methods , Mice , Pancreatic Polypeptide/metabolism , Peritoneum , Rats, Sprague-Dawley
7.
Article En | MEDLINE | ID: mdl-26151373

Both intrinsic and extrinsic factors modulate food intake and glycemia in vertebrates, in part through interactions with hypothalamic neuropeptide Y (NPY) and proopiomelanocortin (POMC) neurons. The objective of this project was to elucidate the effects of ghrelin (GHRL), gastrin-releasing peptide (GRP), cholecystokinin (CCK), glucagon-like peptide (GLP), pancreatic polypeptide (PP), and peptide YY (PYY) on appetite, glycemia, and hypothalamic expression of NPY and POMC in channel catfish. Catfish were injected intraperitoneally with a single peptide at concentrations of either 0 (control), 50, 100, or 200 ng/g body weight (BW), respectively. Fish were allowed to recover for 30 min, and then fed to satiation over 1 h. Feed intake was determined 1h post-feeding. Catfish injected with GHRL at 50 and 100 ng/g BW and GRP at 200 ng/g BW consumed significantly (P<0.05) less feed compared to controls. A tendency (P<0.1) to suppress feed intake was also observed in the 200 ng/g BW GHRL and PP treatments. PYY, CCK, and GLP had no effects on feed intake. Glycemia was not affected by GHRL, GRP, PP, and PYY treatments, but was suppressed by CCK. A tendency toward lower plasma glucose concentrations was observed in fish administered GLP at 50 ng/g BW. Hypothalamic NPY expression was highly variable and not significantly affected by treatment. POMC expression was also variable, but tended to be reduced by the highest concentration of CCK. These results provide new insight into the roles and regulation of gut neuropeptides in catfish appetite and glycemia.


Blood Glucose/metabolism , Eating/drug effects , Ictaluridae/physiology , Neuropeptide Y/genetics , Peptide Hormones/pharmacology , Pro-Opiomelanocortin/genetics , Animals , Cholecystokinin/administration & dosage , Cholecystokinin/pharmacology , Dose-Response Relationship, Drug , Eating/physiology , Gastrin-Releasing Peptide/administration & dosage , Gastrin-Releasing Peptide/pharmacology , Gene Expression/drug effects , Ghrelin/administration & dosage , Ghrelin/pharmacology , Glucagon-Like Peptides/administration & dosage , Glucagon-Like Peptides/pharmacology , Hypothalamus/metabolism , Ictaluridae/blood , Ictaluridae/genetics , Injections, Intraperitoneal , Pancreatic Polypeptide/administration & dosage , Pancreatic Polypeptide/pharmacology , Peptide Hormones/administration & dosage , Peptide YY/administration & dosage , Peptide YY/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
8.
FEBS Lett ; 588(17): 3233-9, 2014 Aug 25.
Article En | MEDLINE | ID: mdl-25019573

Pancreatic polypeptide (PP) is a major agonist for neuropeptide Y4 receptors (NPY4R). While NPY4R has been identified in various tissues, the cells on which it is expressed and its function in those cells has not been clearly delineated. Here we report that NPY4R is present in all somatostatin-containing cells of tissues that we tested, including pancreatic islets, duodenum, hippocampus, and hypothalamus. Its agonism by PP decreases somatostatin secretion from human islets. Mouse embryonic hippocampal (mHippo E18) cells expressed NPY4Rs and their activation by PP consistently decreased somatostatin secretion. Furthermore, central injection of PP in mice induced c-Fos immunoreactivity in somatostatin-containing cells in the hippocampus compared with PBS-injected mice. In sum, our results identify PP as a pivotal modulator of somatostatin secretion.


Pancreatic Polypeptide/pharmacology , Somatostatin/metabolism , Animals , Gene Expression Regulation/drug effects , Hippocampus/cytology , Humans , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Organ Specificity , Receptors, Neuropeptide Y/agonists , Receptors, Neuropeptide Y/metabolism
9.
Am J Physiol Gastrointest Liver Physiol ; 304(4): G437-48, 2013 Feb 15.
Article En | MEDLINE | ID: mdl-23275611

Copper plays an essential role in the function and development of the central nervous system and exocrine pancreas. Dietary copper limitation is known to result in noninflammatory atrophy of pancreatic acinar tissue. Our recent studies have suggested that vagal motoneurons regulate pancreatic exocrine secretion (PES) by activating selective subpopulations of neurons within vagovagal reflexive neurocircuits. We used a combination of in vivo, in vitro, and immunohistochemistry techniques in a rat model of copper deficiency to investigate the effects of a copper-deficient diet on the neural pathways controlling PES. Duodenal infusions of Ensure or casein, as well as microinjections of sulfated CCK-8, into the dorsal vagal complex resulted in an attenuated stimulation of PES in copper-deficient animals compared with controls. Immunohistochemistry of brain stem slices revealed that copper deficiency reduced the number of tyrosine hydroxylase-immunoreactive, but not neuronal nitric oxide synthase- or choline acetyltransferase-immunoreactive, neurons in the dorsal motor nucleus of the vagus (DMV). Moreover, a copper-deficient diet reduced the number of large (>11 neurons), but not small, intrapancreatic ganglia. Electrophysiological recordings showed that DMV neurons from copper-deficient rats are less responsive to CCK-8 or pancreatic polypeptide than are DMV neurons from control rats. Our results demonstrate that copper deficiency decreases efferent vagal outflow to the exocrine pancreas. These data indicate that the combined selective loss of acinar pancreatic tissue and the decreased excitability of efferent vagal neurons induce a deficit in the vagal modulation of PES.


Copper/deficiency , Pancreas/metabolism , Vagus Nerve/physiology , Animals , Caseins/administration & dosage , Dietary Sucrose/pharmacology , Female , Food, Formulated , Male , Motor Neurons/drug effects , Motor Neurons/physiology , Pancreas/innervation , Pancreatic Polypeptide/pharmacology , Rats , Rats, Sprague-Dawley , Sincalide/analogs & derivatives , Sincalide/pharmacology
10.
Neuropeptides ; 47(1): 19-23, 2013 Feb.
Article En | MEDLINE | ID: mdl-22944736

Peripheral injection of pancreatic polypeptide (PP) and peptide YY(3-36) (PYY(3-36)), the hormones released in response to meals, reduce food intake, in which the rank order of the potency is PP>PYY(3-36). These anorectic effects are abolished in abdominal vagotomized rats, suggesting that PP and PYY(3-36) induce anorexia via vagal afferent nerves. However, it is not clear whether PP and PYY(3-36) directly act on vagal afferent neurons. In this study, we examined the effects of PP and PYY(3-36) on cytosolic Ca(2+) concentration ([Ca(2+)](i)) in isolated nodose ganglion neurons of the mouse vagal afferent nerves. At 10(-11)M, PP but not PYY(3-36) recruited a significant population of nodose ganglion neurons into [Ca(2+)](i) increases. PP at 10(-11) to 10(-7) and PYY(3-36) at 10(-10) to 10(-7)M increased [Ca(2+)](i) in a concentration-dependent manner. At submaximal to maximal concentrations of 10(-10) and 10(-8)M, PP increased [Ca(2+)](i) in approximately twice greater population of nodose ganglion neurons than PYY(3-36). Furthermore, the majority of PP-responsive neurons also exhibited [Ca(2+)](i) responses to cholecystokinin-8, a hormone known to induce satiety through activating nodose ganglion neurons. The results demonstrate that PP and PYY(3-36) directly activate nodose ganglion neurons and suggest that the marked effect of PP on cholecystokinin-8-responsive nodose ganglion neurons could be linked to the regulation of feeding.


Calcium Signaling/drug effects , Neurons/drug effects , Neuropeptide Y/pharmacology , Nodose Ganglion/drug effects , Pancreatic Polypeptide/pharmacology , Peptide Fragments/pharmacology , Peptide YY/pharmacology , Animals , Capsaicin/pharmacology , Eating/physiology , Humans , In Vitro Techniques , Male , Mice , Mice, Inbred ICR , Neurons, Afferent/drug effects , Nodose Ganglion/cytology , Potassium Chloride/pharmacology , Rats , Sincalide/metabolism , Vagus Nerve/cytology , Vagus Nerve/drug effects
11.
Peptides ; 39: 6-10, 2013 Jan.
Article En | MEDLINE | ID: mdl-23123314

Pancreatic polypeptide (PP), peptide YY (PYY) and neuropeptide Y (NPY), members of the PP-fold family share a high degree of sequence homology. Nuclear magnetic resonance (NMR) and X-ray crystallography studies have shown these peptides can adopt a tightly organized tertiary structure called the PP-fold, which has long been assumed to be the active structure of this family of peptides. To date, however, no studies have been completed with PYY and PP which confirm if the PP-fold structure is important for their physiological actions. The aim of the study was to test if PYY and PP locked into the PP-fold maintained biological activity. Therefore, we designed and produced analogs of PP and PYY in a cyclic conformation with two cysteine amino acid substitutions at the N-terminus and at position 27. These were oxidized to form a cysteine disulfide bond locking the peptides into the PP-fold structure. Studies demonstrate that the cyclic analogs have both similar in vivo activity to their parent molecules, and affinity for the Y2 and Y4 receptors. Results suggest that the proposed PP and PYY-fold is likely to be their biologically active conformation.


Cystine/chemistry , Eating/drug effects , Pancreatic Polypeptide/chemistry , Peptide YY/chemistry , Peptides, Cyclic/chemistry , Animals , Cystine/chemical synthesis , Food Deprivation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Pancreatic Polypeptide/pharmacology , Peptide YY/pharmacology , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Neuropeptide Y/chemistry
12.
J Neuroendocrinol ; 23(4): 371-80, 2011 Apr.
Article En | MEDLINE | ID: mdl-21251093

Peptide YY (PYY) and pancreatic polypeptide (PP) are two appetite suppressing hormones, released post-prandially from the ileum and pancreas, respectively. PYY(3-36) , the major circulating form of the peptide, is considered to reduce food intake in humans and rodents via high affinity binding to the auto-inhibitory neuropeptide Y receptor Y2R, whereas PP is considered to act through the Y4R. Current evidence indicates the anorexigenic effects of both peptides occur via signalling in the brainstem and arcuate nucleus (ARC) of the hypothalamus. Manganese-enhanced magnetic resonance imaging (MEMRI) has previously been used to track hypothalamic neuronal activity in vivo in response to both nutritional interventions and gut hormone treatment. In the present study, we used MEMRI to demonstrate that s.c. administration of PP results in a significant reduction in signal intensity (SI) in the ARC, ventromedial hypothalamus and paraventricular nucleus of fasted mice. Subcutaneous delivery of PYY(3-36) resulted in a nonsignificant trend towards decreased SI in the hypothalamus of fasted mice. We found no SI change in the area postrema of the brainstem after s.c. injection of either peptide. These differences in hypothalamic SI profile between PP and PYY(3-36) occurred despite both peptides producing a comparable reduction in food intake. These results suggest that separate central pathways control the anorexigenic response for PP and PYY(3-36) , possibly via a differential effect of Y4 receptor versus Y2 receptor signalling. In addition, we performed a series of MEMRI scans at 0-2, 2-4 and 4-6 h post-injection of PYY(3-36) and a potent analogue of the peptide; PYY(3-36) (LT). We recorded a significant reduction in the ARC SI 2-4 h after PYY(3-36) (LT) injection compared to both saline and PYY(3-36) in fasted mice. The physiological differences between PYY(3-36) and its analogue were also observed in the long-term effects on food intake, with PYY(3-36) (LT) producing a more sustained anorexigenic effect. These data suggest that MEMRI can be used to investigate the long-term effects of gut peptide delivery on activity within the hypothalamus and brainstem.


Hypothalamus/cytology , Magnetic Resonance Imaging/methods , Manganese/metabolism , Neurons/metabolism , Pancreatic Polypeptide/metabolism , Peptide YY/metabolism , Animals , Brain Stem/drug effects , Brain Stem/metabolism , Eating/drug effects , Fasting , Humans , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/drug effects , Pancreatic Polypeptide/pharmacology , Peptide Fragments , Peptide YY/pharmacology
13.
J Anim Sci ; 88(6): 2103-7, 2010 Jun.
Article En | MEDLINE | ID: mdl-20190166

The roles of pancreatic polypeptide (PP) have not been determined in ruminant animals. The aim of the present study was to examine the role of PP in the regulation of ghrelin secretion in sheep. Two experiments were conducted using four 2-yr-old Suffolk wethers fed a maintenance diet of alfalfa hay cubes. In Exp. 1, the effects of feeding on blood ghrelin and PP concentrations were examined in scheduled-fed sheep. Blood samples were collected every 10 min from 30 min before to 360 min after feeding. Plasma PP concentrations were transiently increased from the preprandial average value to the values from 30 to 60 min after feeding and gradually decreased (P < 0.05) to stable values from 150 to 180 min. The values from 30 to 60 min were greater (P < 0.05) than those from 150 to 360 min. In contrast, plasma ghrelin concentrations were gradually decreased (P < 0.01) by feeding. The values from 60 to 360 min were less (P < 0.01) than the preprandial average value. In Exp. 2, the effects of continuous PP infusion on ghrelin secretion were examined in feed-deprived sheep. The animals were deprived of feed for 48 h before PP infusion. The PP-treated group intravenously received synthetic bovine PP at a rate of 10 pmol.kg(-1 )of BW.min(-1) for 180 min. Blood samples were collected every 10 min from 30 min before to 180 min after the commencement of PP infusion. Plasma PP concentrations reached a plateau within 30 min after the commencement of PP infusion. Plasma ghrelin concentrations were decreased (P = 0.002, 0.016, 0.007) by PP infusion at 160, 170, and 180 min, respectively. In conclusion, plasma ghrelin and PP concentrations were decreased and increased, respectively, in response to feeding in ruminant animals. Furthermore, PP could depress ghrelin secretion.


Food Deprivation/physiology , Ghrelin/physiology , Pancreatic Polypeptide/pharmacology , Sheep/physiology , Animals , Ghrelin/blood , Ghrelin/metabolism , Linear Models , Male , Pancreatic Polypeptide/blood
14.
Peptides ; 31(6): 1055-61, 2010 Jun.
Article En | MEDLINE | ID: mdl-20338207

Pancreatic polypeptide (PP) belongs to the neuropeptide Y (NPY) family of peptides and is released from pancreatic F cells postprandially. PP functions as a peptide hormone and has been associated with decreased food intake in humans and rodents. Our study describes the effects of PP on feeding behavior in dogs, whose mammalian order (Carnivora) is more distantly related to primates and rodents than these are to each other. Furthermore, obesity is becoming more prevalent in dogs which makes knowledge about their appetite regulation highly relevant. Repeated peripheral administration of physiological doses of PP (three injections of 30 pmol/kg each that were administered within 30 min) to six male beagle dogs prolonged the median time spent eating three servings of food by 19% but resulted in no reduction of food intake. In addition, PP decreased the duration of food-seeking behavior after the first serving by 71%. Thus, a physiological dose of PP seems to decrease both the appetitive and the consummatory drive in dogs.


Appetite Regulation/drug effects , Eating/drug effects , Feeding Behavior/drug effects , Pancreatic Polypeptide/pharmacology , Animals , Behavior, Animal/drug effects , Dogs , Humans , Male , Pancreatic Polypeptide/blood
15.
Neuropeptides ; 44(3): 261-8, 2010 Jun.
Article En | MEDLINE | ID: mdl-20116098

Gut-derived peptides are known to regulate food intake by activating specific receptors in the brain, but the target nuclei and neurons influenced are largely unknown. Here we show that peripherally administered pancreatic polypeptide (PP) stimulates neurons in key nuclei of the hypothalamus critical for appetite and satiety regulation. In the lateral hypothalamic area (LHA), also known as the feeding center, neurons expressing the orexigenic neuropeptide orexin co-localize with the early neuronal activation marker c-Fos upon i.p. injection of PP into mice. In the ventromedial hypothalamus (VMH), also known as the satiety center, neurons activated by PP, as indicated by induction of c-Fos immunoreactivity, express the anorexigenic brain-derived neurotrophic factor (BDNF). Activation of neurons in the LHA and VMH in response to PP occurs via a Y4 receptor-dependent process as it is not seen in Y4 receptor knockout mice. We further demonstrate that in response to i.p. PP, orexin mRNA expression in the LHA is down-regulated, with Y4 receptors being critical for this effect as it is not seen in Y4 receptor knockout mice, whereas BDNF mRNA expression is up-regulated in the VMH in response to i.p. PP in the fasted, but not in the non-fasted state. Taken together these data suggest that PP can regulate food intake by suppressing orexigenic pathways by down-regulation of orexin and simultaneously increasing anorexigenic pathways by up-regulating BDNF.


Brain-Derived Neurotrophic Factor/physiology , Feeding Behavior/physiology , Intracellular Signaling Peptides and Proteins/physiology , Neuropeptides/physiology , Pancreatic Polypeptide/physiology , Receptors, Neuropeptide Y/physiology , Animals , Down-Regulation/physiology , Feeding Behavior/drug effects , Genes, fos/physiology , Hypothalamic Area, Lateral/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orexins , Pancreatic Polypeptide/pharmacology , Receptors, Neuropeptide Y/genetics
16.
Eur J Pharmacol ; 627(1-3): 258-64, 2010 Feb 10.
Article En | MEDLINE | ID: mdl-19818748

Pancreatic polypeptide is released mainly from the pancreas, and is thought to be one of the major endogenous agonists of the neuropeptide Y Y(4) receptor. Pancreatic polypeptide has been shown to stimulate colonic muscle contraction, but whether pancreatic polypeptide has in vivo functional activity with respect to colonic transit is unclear. The present report investigated the effects of pancreatic polypeptide on fecal output as an index of colonic transit as well as intestinal motor activity, using wild-type (WT) and neuropeptide Y Y(4) receptor-deficient (KO) mice. Peripheral administration of pancreatic polypeptide increased fecal weight and caused diarrhea in WT mice in a dose-dependent manner (0.01-3mg/kg s.c.). Pancreatic polypeptide-induced increases in fecal weight and diarrhea completely disappeared in KO mice, while basal fecal weights did not differ between WT and KO mice. In longitudinal and circular muscles of mouse isolated colon, pancreatic polypeptide (0.01-1 microM) increased basal tone and frequency of spontaneous contraction in WT mice, but not in KO mice. Atropine did not affect pancreatic polypeptide-induced fecal output or increase in colonic muscle tone, indicating that the actions of pancreatic polypeptide are not mediated through cholinergic mechanisms. The present findings demonstrate that pancreatic polypeptide enhances colonic contractile activity and fecal output through neuropeptide Y Y(4) receptor, and a neuropeptide Y Y(4) receptor agonist might offer a novel therapeutic approach to ameliorate constipation.


Colon/drug effects , Colon/physiology , Feces , Muscle Contraction/drug effects , Pancreatic Polypeptide/pharmacology , Receptors, Neuropeptide Y/metabolism , Acetylcholine/pharmacology , Animals , Atropine/pharmacology , Colon/metabolism , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Ileum/drug effects , Ileum/physiology , In Vitro Techniques , Intestinal Mucosa/metabolism , Intestines/drug effects , Male , Mice , Mice, Inbred C57BL , Receptors, Neuropeptide Y/deficiency , Receptors, Neuropeptide Y/genetics , Substance P/pharmacology , Water/metabolism
17.
PLoS One ; 4(12): e8488, 2009 Dec 30.
Article En | MEDLINE | ID: mdl-20041129

BACKGROUND: Pancreatic polypeptide (PP) is a potent anti-obesity agent known to inhibit food intake in the absence of nausea, but the mechanism behind this process is unknown. METHODOLOGY/PRINCIPAL FINDINGS: Here we demonstrate that in response to i.p. injection of PP in wild type but not in Y4 receptor knockout mice, immunostaining for the neuronal activation marker c-Fos is induced specifically in neurons of the nucleus tractus solitarius and the area postrema in the brainstem, notably in cells also showing immunostaining for tyrosine hydroxylase. Importantly, strong c-Fos activation is also detected in the arcuate nucleus of the hypothalamus (ARC), particularly in neurons that co-express alpha melanocyte stimulating hormone (alpha-MSH), the anorexigenic product of the proopiomelanocortin (POMC) gene. Interestingly, other hypothalamic regions such as the paraventricular nucleus, the ventromedial nucleus and the lateral hypothalamic area also show c-Fos induction after PP injection. In addition to c-Fos activation, PP injection up-regulates POMC mRNA expression in the ARC as detected by in situ hybridization. These effects are a direct consequence of local Y4 signaling, since hypothalamus-specific conditional Y4 receptor knockout abolishes PP-induced ARC c-Fos activation and blocks the PP-induced increase in POMC mRNA expression. Additionally, the hypophagic effect of i.p. PP seen in wild type mice is completely absent in melanocortin 4 receptor knockout mice. CONCLUSIONS/SIGNIFICANCE: Taken together, these findings show that PP reduces food intake predominantly via stimulation of the anorexigenic alpha-MSH signaling pathway, and that this effect is mediated by direct action on local Y4 receptors within the ARC, highlighting a potential novel avenue for the treatment of obesity.


Arcuate Nucleus of Hypothalamus/metabolism , Feeding Behavior/drug effects , Melanocortins/metabolism , Pancreatic Polypeptide/pharmacology , Receptors, Neuropeptide Y/metabolism , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/enzymology , Brain Stem/cytology , Brain Stem/drug effects , Brain Stem/metabolism , Energy Metabolism/drug effects , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Male , Mice , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptor, Melanocortin, Type 4/metabolism , Receptors, Neuropeptide Y/agonists , Signal Transduction/drug effects , alpha-MSH/metabolism
18.
Neuroscience ; 158(4): 1717-30, 2009 Feb 18.
Article En | MEDLINE | ID: mdl-19121371

There is growing evidence that neuropeptide Y (NPY) acting through Y1 and Y2 receptors has a prominent role in modulating anxiety- and depression-like behavior in rodents. However, a role of other Y-receptors like that of Y4 receptors in this process is poorly understood. We now investigated male Y2, Y4 single and Y2/Y4 double knockout mice in behavioral paradigms for changes in motor activity, anxiety and depression-like behavior. Motor activity was increased in Y2, Y4 and Y2/Y4 knockout mice under changing and stressful conditions, but not altered in a familiar environment. Y4 and Y2 knockout mice revealed an anxiolytic phenotype in the light/dark test, marble burying test and in stress-induced hyperthermia, and reduced depression-like behavior in the forced swim and tail suspension tests. In Y2/Y4 double knockout mice, the response in the light/dark test and in the forced swim test was further enhanced compared with Y4 and Y2 knockout mice, respectively. High levels of Y4 binding sites were observed in brain stem nuclei including nucleus of solitary tract and area postrema. Lower levels were found in the medial amygdala and hypothalamus. Peripheral administration of pancreatic polypeptide (PP) induced Y4 receptor-dependent c-Fos expression in brain stem, hypothalamus and amygdala. PP released peripherally from the pancreas in response to food intake, may act not only as a satiety signal but also modulate anxiety-related locomotion.


Depression/genetics , Depression/physiopathology , Exploratory Behavior/physiology , Motor Activity/genetics , Receptors, Neuropeptide Y/deficiency , Amygdala/metabolism , Animals , Autoradiography/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Hindlimb Suspension/methods , Hyperthermia, Induced , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Polypeptide/pharmacology , Protein Binding , Proto-Oncogene Proteins c-fos/metabolism , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Swimming/physiology
19.
Eur J Pharmacol ; 601(1-3): 118-23, 2008 Dec 28.
Article En | MEDLINE | ID: mdl-18851959

The present study investigated the mechanisms of vasodilatation of the human pancreatic polypeptide [cPP(1-7), NPY(19-23),Ala(31),Aib(32),Gln(34)]hPP (hPP) in mesenteric small arteries from Wistar Kyoto rats (WKY) and spontaneously hypertensive rats (SHR). The arteries were isolated and mounted in microvascular myographs for isometric tension recording. In vasopressin-contracted preparations with endothelium from WKY rats, hPP evoked concentration-dependent relaxations with maximal responses of 50+/-2% (n=5). hPP relaxation was reduced by endothelial cell removal and abolished in the presence of a nitric oxide (NO) synthase inhibitor, N(G)-nitro-L-arginine-methylester (L-NAME). hPP relaxation was blunted in segments with endothelium, and absent in segments without endothelium from SHR. The combined neuropeptide Y(1)- and Y(4)-receptor antagonist, GR23118 (Ile-Glu-Pro-Dpr-Tyr-Arg-Leu-Arg-Tyr-CONH(2)), and the neuropeptide Y(1) receptor antagonist, BIBP3226 ((R) -N2-(diphenylacetyl)-N-[(4-hydroxyphenyl)-methyl]-arginineamide), inhibited hPP-induced vasodilatation. Calcitonin gene-related peptide (CGRP) relaxation was reduced in arteries from SHR compared to WKY. The CGRP receptor antagonist, CGRP (8-37), antagonized vasodilatation induced by CGRP and rightward shifted concentration-response curves for hPP in arteries from WKY rats. There were no differences in nerves immunoreactive for CGRP in arteries from SHR compared to WKY rats. In contrast to neuropeptide Y which evokes contraction by activation of neuropeptide Y(1) and Y(2) receptors, the present results suggest hPP evokes relaxation of mesenteric small arteries by activation of prejunctional neuropeptide Y(1)-like receptors localized in CGRP-containing nerves followed by release of CGRP and of endothelium-derived NO. hPP relaxation is blunted in arteries from SHR probably as a consequence of endothelial cell dysfunction leading to reduced efficacy of CGRP.


Mesenteric Arteries/drug effects , Neuropeptide Y/pharmacology , Pancreatic Polypeptide/pharmacology , Vasodilation/drug effects , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/pharmacology , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Female , Humans , Mesenteric Arteries/metabolism , Neuropeptide Y/metabolism , Nitric Oxide/metabolism , Pancreatic Polypeptide/administration & dosage , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, Calcitonin Gene-Related Peptide/drug effects , Receptors, Calcitonin Gene-Related Peptide/metabolism , Receptors, Neuropeptide Y/drug effects , Receptors, Neuropeptide Y/metabolism , Vasopressins/pharmacology
20.
Int J Obes (Lond) ; 32(11): 1712-5, 2008 Nov.
Article En | MEDLINE | ID: mdl-18779824

Pancreatic polypeptide (PP) has been shown to inhibit food intake in both rodents and humans. Acute peripheral administration of PP increases oxygen consumption in obese mice. To further investigate the function of PP in the modulation of energy balance, we examined its effects on spontaneous locomotor activity and food intake in mice by using a 24-chamber Comprehensive Laboratory Animal Monitoring System. Effects of intraperitoneal (i.p.) administration of PP on spontaneous locomotor activity were measured using the optical beam technique. Administration of PP dose dependently inhibited cumulative food intake. The inhibition remained significant for up to 6, 17 and 36 h at doses of 30, 100 and 300 nmol kg(-1) PP, respectively. At 10 nmol kg(-1), PP increased locomotor activity (cumulative beam breaks) significantly for 4 h following administration without affecting food intake and at 30 nmol kg(-1), PP increased locomotor activity by 79% compared with the controls for 5 h post injection. However, at 100 and 300 nmol kg(-1), PP had no significant effect on locomotor activity. This study shows for the first time that PP increases spontaneous locomotor activity in mice.


Motor Activity/drug effects , Pancreatic Polypeptide/pharmacology , Animals , Body Weight , Eating/drug effects , Eating/physiology , Energy Metabolism , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Motor Activity/physiology , Satiation/physiology
...