Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Physiol Rep ; 4(10)2016 May.
Article in English | MEDLINE | ID: mdl-27207785

ABSTRACT

Glucagon-like peptide-1 (GLP-1), peptide YY (3-36) [PYY(3-36)], amylin, ghrelin, insulin, and leptin are thought to act as hormonal signals from periphery to brain to control food intake. Here, we determined the effects of solid-phase extraction of plasma in measuring these hormones in blood of lean and diet-induced obese rats. Individual enzyme-linked immunoassays and a multiplex assay were used to measure active GLP-1, total PYY, active amylin, active ghrelin, insulin, leptin, and total GIP in response to (1) addition of known amounts of the peptides to lean and obese plasma, (2) a large meal in lean and obese rats, and (3) intravenous infusions of anorexigenic doses of GLP-1, PYY(3-36), amylin, and leptin in lean rats. Extraction of lean and obese plasma prior to assays produced consistent recoveries across assays for GLP-1, PYY, amylin, ghrelin, and insulin, reflecting losses inherent to the extraction procedure. Plasma extraction prior to assays generally revealed larger meal-induced changes in plasma GLP-1, PYY, amylin, ghrelin, and insulin in lean and obese rats. Plasma extraction and the multiplex assay were used to compare plasma levels of GLP-1, PYY, and amylin after a large meal with plasma levels produced by IV infusions of anorexigenic doses of GLP-1, PYY(3-36), and amylin. Infusions produced dose-dependent increases in plasma peptide levels, which were well above their postprandial levels. These results do not support the hypothesis that postprandial plasma levels of GLP-1, PYY(3-36), and amylin are sufficient to decrease food intake by an endocrine mechanism.


Subject(s)
Diet, High-Fat/adverse effects , Eating/physiology , Fasting/blood , Gastrointestinal Hormones/blood , Obesity/blood , Solid Phase Extraction/methods , Thinness/blood , Animals , Eating/drug effects , Gastrointestinal Hormones/administration & dosage , Infusions, Intravenous , Male , Obesity/chemically induced , Rats
2.
Am J Physiol Endocrinol Metab ; 307(8): E619-29, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25117406

ABSTRACT

Cholecystokinin (CCK)-induced suppression of feeding is mediated by vagal sensory neurons that are destroyed by the neurotoxin capsaicin (CAP). Here we determined whether CAP-sensitive neurons mediate anorexic responses to intravenous infusions of gut hormones peptide YY-(3-36) [PYY-(3-36)] and glucagon-like peptide-1 (GLP-1). Rats received three intraperitoneal injections of CAP or vehicle (VEH) in 24 h. After recovery, non-food-deprived rats received at dark onset a 3-h intravenous infusion of CCK-8 (5, 17 pmol·kg⁻¹·min⁻¹), PYY-(3-36) (5, 17, 50 pmol·kg⁻¹·min⁻¹), or GLP-1 (17, 50 pmol·kg⁻¹·min⁻¹). CCK-8 was much less effective in reducing food intake in CAP vs. VEH rats. CCK-8 at 5 and 17 pmol·kg⁻¹·min⁻¹ reduced food intake during the 3-h infusion period by 39 and 71% in VEH rats and 7 and 18% in CAP rats. In contrast, PYY-(3-36) and GLP-1 were similarly effective in reducing food intake in VEH and CAP rats. PYY-(3-36) at 5, 17, and 50 pmol·kg⁻¹·min⁻¹ reduced food intake during the 3-h infusion period by 15, 33, and 70% in VEH rats and 13, 30, and 33% in CAP rats. GLP-1 at 17 and 50 pmol·kg⁻¹·min⁻¹ reduced food intake during the 3-h infusion period by 48 and 60% in VEH rats and 30 and 52% in CAP rats. These results suggest that anorexic responses to PYY-(3-36) and GLP-1 are not primarily mediated by the CAP-sensitive peripheral sensory neurons (presumably vagal) that mediate CCK-8-induced anorexia.


Subject(s)
Anorexia/physiopathology , Cholecystokinin/metabolism , Disease Models, Animal , Intestinal Mucosa/innervation , Intestine, Small/innervation , Neuritis/physiopathology , Neurons, Afferent/metabolism , Peptide Fragments/metabolism , Animals , Anorexia/metabolism , Anorexia/prevention & control , Behavior, Animal/drug effects , Capsaicin/administration & dosage , Capsaicin/toxicity , Cholecystokinin/administration & dosage , Energy Intake/drug effects , Feeding Behavior/drug effects , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/metabolism , Infusions, Intravenous , Injections, Intraperitoneal , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiopathology , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestine, Small/physiopathology , Male , Neuritis/chemically induced , Neuritis/metabolism , Neurons, Afferent/drug effects , Peptide Fragments/administration & dosage , Peptide YY/administration & dosage , Peptide YY/metabolism , Rats , Vagus Nerve/drug effects , Vagus Nerve/metabolism , Vagus Nerve/physiopathology , Vagus Nerve Diseases/chemically induced , Vagus Nerve Diseases/metabolism , Vagus Nerve Diseases/physiopathology
3.
Am J Physiol Endocrinol Metab ; 304(9): E944-50, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23482449

ABSTRACT

Peptide YY(3-36) [PYY(3-36)] is postulated to act as a hormonal signal from gut to brain to inhibit food intake. PYY(3-36) potently reduces food intake when administered systemically or into the brain. If action of endogenous PYY(3-36) is necessary for normal satiation to occur, then pharmacological blockade of its receptors should increase food intake. Here, we determined the effects of iv infusion of Y1, Y2, and Y5 receptor antagonists (BIBP 3226, BIIE 0246, CGP 71683) during the first 3 h of the dark period on food intake in non-food-deprived rats. Our results showed that 1) Y2 receptor blockade reversed the anorexic response to iv infusion of PYY(3-36) but did not increase food intake when administered alone; 2) Y1 and Y5 receptor antagonists neither attenuated PYY(3-36)-induced anorexia nor altered food intake when given alone; and 3) Y2 receptor blockade attenuated anorexic responses to gastric infusions of casein hydrolysate and long-chain triglycerides, but not maltodextrin. Previous work showed that Y2 antagonist BIIE 0246 does not penetrate the blood-brain barrier. Together, these results support the hypothesis that gut PYY(3-36) action at Y2 receptors peripheral to the blood brain barrier plays an essential role in mediating satiety responses to gastric delivery of protein and long-chain triglycerides, but not polysaccharide.


Subject(s)
Peptide Fragments/physiology , Peptide YY/physiology , Satiety Response/physiology , Animals , Anorexia/psychology , Cholecystokinin/physiology , Eating/drug effects , Hunger/physiology , Intubation, Gastrointestinal , Islet Amyloid Polypeptide/physiology , Male , Polysaccharides/pharmacology , Protein Hydrolysates/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Triglycerides/pharmacology
4.
Am J Physiol Endocrinol Metab ; 302(12): E1576-85, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22510712

ABSTRACT

Weight loss in obese humans produces a relative leptin deficiency, which is postulated to activate potent orexigenic and energy conservation mechanisms to restrict weight loss and promote weight regain. Here we determined whether leptin replacement alone or with GLP-1 receptor agonist exendin-4 attenuates weight regain or promotes greater weight loss in weight-reduced diet-induced obese (DIO) rats. Forty percent restriction in daily intake of a high-fat diet in DIO rats for 4 wk reduced body weight by 12%, body fat by 29%, and plasma leptin by 67% and normalized leptin sensitivity. When food restriction ended, body weight, body fat, and plasma leptin increased rapidly. Daily administration of leptin [3-h intraperitoneal (ip) infusions (4 nmol·kg(-1)·h(-1))] at onset and end of dark period for 3 wk did not attenuate hyperphagia and weight regain, nor did it affect mean daily meal sizes or meal numbers. Exendin-4 (50 pmol·kg(-1)·h(-1)) infusions during the same intervals prevented postrestriction hyperphagia and weight regain by normalizing meal size. Coadministration of leptin and exendin-4 did not reduce body weight more than exendin-4 alone. Instead, leptin began to attenuate the inhibitory effects of exendin-4 on food intake, meal size, and weight regain by the end of the second week of administration. Plasma leptin in rats receiving leptin was sevenfold greater than in rats receiving vehicle and 17-fold greater than in rats receiving exendin-4. Together, these results do not support the hypothesis that leptin replacement alone or with exendin-4 attenuates weight regain or promotes greater weight loss in weight-reduced DIO rats.


Subject(s)
Eating/drug effects , Leptin/pharmacology , Obesity/diet therapy , Peptides/pharmacology , Venoms/pharmacology , Weight Gain/drug effects , Weight Loss/physiology , Animals , Body Composition/physiology , Body Fat Distribution , Body Weight/physiology , Caloric Restriction , Dose-Response Relationship, Drug , Exenatide , Glucagon-Like Peptide-1 Receptor , Hyperphagia/psychology , Leptin/blood , Male , Obesity/psychology , Rats , Rats, Sprague-Dawley , Receptors, Glucagon/agonists
5.
Peptides ; 32(4): 770-5, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21262301

ABSTRACT

The gut hormone peptide YY(3-36)-amide [PYY(3-36)-NH(2)] is significantly more potent than PYY(1-36)-NH(2) in reducing food intake in rats and humans. Other Gly-extended and Ser(13)-phosphorylated PYY forms have been detected or predicted based upon known cellular processes of PYY synthesis and modification. Here we compared the effects of 3-h IV infusion of PYY(1-36)-NH(2), PYY(3-36)-NH(2), PYY(1-36)-Gly-OH, PYY(3-36)-Gly-OH, Ser(13)(PO(3))-PYY(1-36)-NH(2), Ser(13)(PO(3))-PYY(3-36)-NH(2), Ser(13)(PO(3))-PYY(1-36)-Gly-OH, and Ser(13)(PO(3))-PYY(3-36)-Gly-OH during the early dark period on food intake in freely feeding rats. PYY(3-36)-NH(2) and Ser(13)(PO(3))-PYY(3-36)-NH(2) reduced food intake similarly at 50 pmol/kg/min, while only PYY(3-36)-NH(2) reduced food intake at 15 pmol/kg/min. PYY(1-36)-NH(2) and Ser(13)(PO(3))-PYY(1-36)-NH(2) reduced food intake similarly at 50 and 150 pmol/kg/min. In contrast, PYY(1-36)-Gly-OH, PYY(3-36)-Gly-OH, Ser(13)(PO(3))-PYY(3-36)-Gly-OH, and Ser(13)(PO(3))-PYY(1-36)-Gly-OH had no effect on food intake at doses of 50 or 150 pmol/kg/min. Taken together, these results indicate that (i) PYY(3-36)-NH(2) is significantly more potent than PYY(1-36)-NH(2) in reducing food intake, (ii) Gly-extended forms of PYY are significantly less potent than non-extended forms, and (iii) Ser(13)-phosphorylation of PYY(3-36)-NH(2) decreases the anorexigenic potency PYY(3-36)-NH(2), but not PYY(1-36)-NH(2). Thus, PYY(3-36)-NH(2) appears to be the most potent PYY form for reducing food intake in rats.


Subject(s)
Feeding Behavior/drug effects , Glycine/chemistry , Peptide YY/pharmacology , Serine/chemistry , Animals , Peptide YY/chemistry , Phosphorylation , Rats
6.
Obesity (Silver Spring) ; 19(1): 121-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20559304

ABSTRACT

Significant weight loss following Roux-en-Y gastric bypass surgery (RYGB) in obese humans correlates with enhanced secretion of anorexigenic gut hormones glucagon-like peptide-1 (GLP-1) and peptide YY(3-36) (PYY(3-36)). Our aim here was to identify a dosing strategy for intraperitoneal (IP) infusion of GLP-1 homologue exendin-4 alone and with PYY(3-36) that produces a sustained reduction in daily food intake and body weight in diet-induced obese (DIO) rats. We tested 12 exendin-4 strategies over 10 weeks. Exendin-4 infused during the first and last 3 h of the dark period at 15-20 pmol/h (0.15 nmol/kg/day) produced a sustained 24 ± 1% reduction in daily food intake for 17 days, and decreased body weight by 7%. In a separate group of DIO rats, none of seven dosing strategies combining exendin-4 and PYY(3-36) produced a similar reduction in daily food intake for >10 days. The subsequent decline in efficacies of exendin-4 alone and with PYY(3-36) on food intake and body weight in each experiment suggested possible receptor downregulation and tolerance to treatments. However, when treatments were discontinued for 1 day following losses in efficacies, daily food intake significantly increased. Together, these results demonstrate that (i) intermittent IP infusion of a low dose of exendin-4 can produce a relatively prolonged reduction in daily food intake and body weight in DIO rats, (ii) co-infusion of exendin-4 and PYY(3-36) does not further prolong this response, and (iii) activation of an orexigenic mechanism gradually occurs to counteract the inhibitory effects of exendin-4 alone and with PYY(3-36) on food intake and body weight.


Subject(s)
Body Weight/drug effects , Eating/drug effects , Obesity/physiopathology , Peptide YY/administration & dosage , Peptides/administration & dosage , Peptides/pharmacology , Venoms/administration & dosage , Venoms/pharmacology , Animals , Body Weight/physiology , Diet/adverse effects , Dose-Response Relationship, Drug , Drug Combinations , Drug Evaluation, Preclinical , Eating/physiology , Exenatide , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Male , Obesity/drug therapy , Obesity/etiology , Peptide Fragments , Peptide YY/pharmacology , Rats , Rats, Sprague-Dawley , Weight Loss/drug effects
7.
Regul Pept ; 165(2-3): 151-7, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20615437

ABSTRACT

We measured molecular forms of PYY in the distal half of rat small intestine using a new method for tissue extraction, three sequential reverse phase chromatography steps, and PYY radioimmunoassay and mass spectrometry to measure their levels. The extraction method called RAPID, developed to minimize artifactual degradation of PYY during tissue extraction and sample preparation, uses Reduced temperature, Acidified buffer, Peptidase inhibitors, Isotopically enriched mass spectrometry standards, and Dilution to inhibit and monitor endogenous peptide degradation during tissue processing. Synthetic peptides [PYY(1-36)-NH(2), PYY(3-36)-NH(2), PYY(1-36)-Gly-OH, and PYY(3-36)-Gly-OH] selectively enriched with (13)C(3)-alanine were added as internal standards to the extraction buffer. By collecting mass spectra rather than multiple-reaction-monitoring (MRM) profiles, we simultaneously screen for any PYY forms that were present in the immunoreactive fractions. PYY(1-36)-NH(2), PYY(3-36)-NH(2), PYY(1-36)-Gly-OH, and PYY(3-36)-Gly-OH were identified and quantified at 64.3±4.5, 6.1±0.9, 0.9±0.1, and <0.3pmol/g of tissue, respectively (n=3). Thus, we found that in rat distal small intestine proPYY is processed to PYY(1-36)-NH(2) with little conversion to PYY(3-36)-NH(2). These data suggest that production of PYY(3-36)-NH(2) (a form with greater potency than PYY(1-36)-NH(2) for inhibition of feeding and gastric emptying) occurs after the peptide leaves its cell of synthesis by enzymatic action in the circulation.


Subject(s)
Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Peptide YY/metabolism , Animals , Male , Mass Spectrometry , Radioimmunoassay , Rats , Rats, Sprague-Dawley
8.
Am J Physiol Regul Integr Comp Physiol ; 295(2): R449-58, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18550871

ABSTRACT

Chronic administration of anorexigenic substances to experimental animals by injections or continuous infusion typically produces either no effect or a transient reduction in food intake and body weight. Our aim here was to identify an intermittent dosing strategy for intraperitoneal infusion of peptide YY(3-36) [PYY(3-36)] that produces a sustained reduction in daily food intake and adiposity in diet-induced obese rats. Rats (665+/-10 g body wt, 166+/-7 g body fat) with intraperitoneal catheters tethered to infusion swivels had free access to a high-fat diet. Vehicle-treated rats (n=23) had relatively stable food intake, body weight, and adiposity during the 9-wk test period. None of 15 PYY(3-36) dosing regimens administered in succession to a second group of rats (n=22) produced a sustained 15-25% reduction in daily food intake for >5 days, although body weight and adiposity were reduced across the 9-wk period by 12% (594+/-15 vs. 672+/-15 g) and 43% (96+/-7 vs. 169+/-9 g), respectively. The declining inhibitory effect of PYY(3-36) on daily food intake when the interinfusion interval was >or=3 h appeared to be due in part to an increase in food intake between infusions. The declining inhibitory effect of PYY(3-36) on daily food intake when the interinfusion interval was <3 h suggested possible receptor downregulation and tolerance to frequent PYY(3-36) administration; however, food intake significantly increased when PYY(3-36) treatments were discontinued for 1 day following apparent loss in treatment efficacies. Together, these results demonstrate the development of a potent homeostatic response to increase food intake when PYY(3-36) reduces food intake and energy reserves in diet-induced obese rats.


Subject(s)
Adiposity/drug effects , Appetite Depressants/administration & dosage , Behavior, Animal/drug effects , Body Weight/drug effects , Eating/drug effects , Obesity/drug therapy , Peptide YY/administration & dosage , Animals , Dietary Fats , Disease Models, Animal , Drug Administration Schedule , Homeostasis , Infusions, Parenteral , Male , Obesity/etiology , Obesity/physiopathology , Peptide Fragments , Rats , Rats, Sprague-Dawley , Time Factors
9.
Regul Pept ; 151(1-3): 61-70, 2008 Nov 29.
Article in English | MEDLINE | ID: mdl-18501442

ABSTRACT

We purified and identified the peptide YY (PYY) forms present and determined their levels from a portion of the canine ileum directly adjacent to the cecum by a new extraction method designed to prevent and evaluate degradation of endogenous peptides. We used three reverse phase chromatography steps with radioimmunoassay of fractions for PYY-like-immunoreactivity (PYY-LI). The purified fractions underwent intact protein/peptide mass spectrometry identification and sequencing (i.e. "top-down" MS analysis). This analysis confirmed the identity of a new form of PYY, PYY(1-36)-Gly, which co-elutes with PYY(1-36)-NH(2) through all three of separation steps used. The PYY(1-36)-Gly form represents approximately 20% of the total PYY found in this region of the canine intestine. In addition, we also found that the PYY(3-36)-NH(2) form represents 6% of the total PYY in the canine ileo-cecal junction. The physiological implication of the Gly-extended form of PYY(1-36) warrants further investigation.


Subject(s)
Ileum/chemistry , Peptide YY/chemistry , Peptide YY/isolation & purification , Amino Acid Sequence , Animals , Dogs , Ileum/anatomy & histology , Mass Spectrometry , Molecular Sequence Data , Molecular Structure , Peptide YY/genetics , Radioimmunoassay , Sequence Homology, Amino Acid , Tissue Distribution
10.
Am J Physiol Regul Integr Comp Physiol ; 293(5): R1798-808, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17761508

ABSTRACT

Chronic administration of anorexigenic substances to experimental animals by injections or continuous infusion typically produces no effect or a transient reduction in daily food intake and body weight. Our aim was to identify an intermittent dosing strategy for intraperitoneal infusion of salmon calcitonin (sCT), a homolog of amylin that produces a sustained 25-35% reduction in daily food intake and adiposity in diet-induced obese rats. Rats (649 +/- 10 g body wt, 27 +/- 1% body fat), with intraperitoneal catheters tethered to infusion swivels, had free access to a 45% fat diet. Food intake, body weight, and adiposity during the 7-wk test period were relatively stable in the vehicle-treated rats (n = 16). None of 10 sCT dosing regimens administered in succession to a second group of rats (n = 18) produced a sustained 25-35% reduction in daily food intake for >5 days, although body weight and adiposity were reduced by 9% (587 +/- 12 vs. 651 +/- 14 g) and 22% (20.6 +/- 1.2 vs. 26.5 +/- 1.1%), respectively, across the 7-wk period. The declining inhibitory effect of sCT on daily food intake with the 6-h interinfusion interval appeared to be due in part to an increase in food intake between infusions. The declining inhibitory effect of sCT on daily food intake with the 2- to 3-h interinfusion interval suggested possible receptor downregulation and tolerance to frequent sCT administration; however, food intake increased dramatically when sCT was discontinued for 1 day after apparent loss of treatment efficacy. Together, these results demonstrate the activation of a potent homeostatic response to increase food intake when sCT reduces food intake and energy reserves in diet-induced obese rats.


Subject(s)
Adiposity/drug effects , Calcitonin/pharmacology , Eating/drug effects , Obesity/drug therapy , Amyloid/metabolism , Animals , Body Weight/drug effects , Calcitonin/administration & dosage , Diet , Infusions, Parenteral , Islet Amyloid Polypeptide , Male , Obesity/physiopathology , Rats , Rats, Sprague-Dawley
11.
Am J Physiol Regul Integr Comp Physiol ; 293(1): R39-46, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17428898

ABSTRACT

Peptide YY(3-36) [PYY(3-36)] is a gut-brain peptide that decreases food intake when administered by intravenous infusion to lean and obese humans and rats. However, chronic administration of PYY(3-36) by osmotic minipump to lean and obese rodents produces only a transient reduction in daily food intake and weight gain. It has recently been shown that 1-h intravenous infusions of PYY(3-36) every other hour for 10 days produced a sustained reduction in daily food intake, body weight, and adiposity in lean rats. Here, we determined whether intermittent delivery of PYY(3-36) can produce a similar response in diet-induced obese rats. During a 21-day period, obese rats (body fat >25%) received twice daily intraperitoneal infusion of vehicle (n = 18) or PYY(3-36) (n = 24) during hours 1-3 and 7-9 of the dark period. Rats had free access to both a 45% fat solid diet and a 29% fat liquid diet; intakes were determined from continuous computer recording of changes in food container weights. To sustain a 15-25% reduction in daily caloric intake, the initial PYY(3-36) dose of 30 pmol.kg(-1).min(-1) was reduced to 10 pmol.kg(-1).min(-1) on day 10 and then increased to 17 pmol.kg(-1).min(-1) on day 13. This dosing strategy produced a sustained reduction in daily caloric intake of 11-32% and prevented body weight gain (8 +/- 6 vs. 51 +/- 11 g) and fat deposition (4.4 +/- 7.6 vs. 41.0 +/- 12.8 g). These results indicate that intermittent intraperitoneal infusion of PYY(3-36) can produce a sustained reduction in food intake and adiposity in diet-induced obese rodents consuming palatable high-fat foods.


Subject(s)
Adiposity/drug effects , Eating/drug effects , Obesity/drug therapy , Peptide YY/pharmacology , Animals , Body Weight/drug effects , Diet , Dose-Response Relationship, Drug , Energy Intake/drug effects , Infusions, Intravenous , Infusions, Parenteral , Male , Obesity/physiopathology , Peptide Fragments , Peptide YY/administration & dosage , Rats , Rats, Sprague-Dawley , Weight Gain/drug effects
12.
Diabetes ; 55(11): 3038-46, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17065340

ABSTRACT

Ghrelin stimulates, while glucagon-like peptide-1 (GLP-1) and peptide YY(3-36) [PYY(3-36)] inhibit, food intake and gastric emptying in rats. We determined the dose-dependent effects of a 3-h intravenous infusion of ghrelin at dark onset on food intake in freely feeding rats, and on the inhibitory effects of intravenous infusion of GLP-1 and PYY(3-36) on food intake and gastric emptying. Ghrelin (150 pmol x kg(-1) x min(-1)) stimulated food intake by 28% during the infusion period primarily by increasing meal frequency; doses of 15 and 50 pmol x kg(-1) x min(-1) had no effect. GLP-1 (15 pmol x kg(-1) x min(-1)) inhibited food intake by 35-54%; coinfusion of ghrelin at 50 and 150 pmol x kg(-1) x min(-1) attenuated this effect by 60 and 64%, respectively. PYY(3-36) (15 pmol x kg(-1) x min(-1)) inhibited food intake by 32%; ghrelin at 15 and 50 pmol x kg(-1) x min(-1) attenuated this effect by 54 and 74%, respectively. A 20-min intravenous infusion of ghrelin (15-150 pmol x kg(-1) x min(-1)) attenuated GLP-1-and PYY(3-36)-induced inhibition of gastric emptying of saline by 6-29%. Thus, intravenous infusion of ghrelin during the early dark period stimulates food intake in freely feeding rats by increasing meal frequency, and similar doses of ghrelin attenuate gastric emptying and feeding responses to GLP-1 and PYY(3-36). These results suggest that ghrelin may stimulate food intake in part by attenuating the inhibitory effects of GLP-1 and PYY(3-36) on gastric emptying and food intake.


Subject(s)
Eating/drug effects , Gastric Emptying/drug effects , Glucagon-Like Peptide 1/pharmacology , Peptide Hormones/pharmacology , Peptide YY/pharmacology , Animals , Ghrelin , Glucagon-Like Peptide 1/antagonists & inhibitors , Infusions, Intravenous , Kinetics , Male , Peptide Fragments , Peptide Hormones/drug effects , Peptide YY/antagonists & inhibitors , Rats , Rats, Sprague-Dawley
13.
Peptides ; 27(12): 3193-201, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16962209

ABSTRACT

We used a conditioned taste aversion test to assess whether PYY(3-36) reduces food intake by producing malaise. Two-hour IV infusion of PYY(3-36) (8, 15, and 30 pmol/kg/min) at dark onset in non-food-deprived rats produced a dose-dependent inhibition of feeding and a conditioned aversion to the flavored chow paired with PYY(3-36) infusion. In food-deprived rats, PYY(3-36) at 2 and 4 pmol/kg/min inhibited intake of a flavored saccharin solution without producing conditioned taste aversion, whereas higher doses (8 and 15 pmol/kg/min) inhibited saccharin intake and produced taste aversion. These results suggest that anorexic doses of PYY(3-36) may produce a dose-dependent malaise in rats, which is similar to that reported for PYY(3-36) infusion in humans. Previous studies have shown that PYY(3-36) potently inhibits gastric emptying, and that gut distention can produce a conditioned taste aversion. Thus, PYY(3-36) may produce conditioned taste aversion in part by slowing gastric emptying.


Subject(s)
Avoidance Learning/drug effects , Peptide Fragments/physiology , Peptide YY/physiology , Taste/physiology , Animals , Conditioning, Operant , Dose-Response Relationship, Drug , Infusions, Intravenous , Male , Peptide Fragments/administration & dosage , Peptide YY/administration & dosage , Rats , Rats, Sprague-Dawley
14.
Am J Physiol Regul Integr Comp Physiol ; 290(2): R298-305, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16210414

ABSTRACT

The gut hormone peptide YY(3-36) [PYY(3-36)] decreases food intake when administered by intravenous infusion to lean and obese humans and rats. Whether chronic administration of PYY(3-36) produces a sustained reduction in food intake and adiposity is the subject of intense debate. Batterham et al. (R. L. Batterham, M. A. Cowley, C. J. Small, H. Herzog, M. A. Cohen, C. L. Dakin, A. M. Wren, A. E. Brynes, M. J. Low, M. A. Ghatei, R. D. Cone, and S. R. Bloom. Nature 418: 650-654, 2002) first reported that PYY(3-36) reduces food intake and weight gain in rats when injected into the peritoneal cavity twice daily for 7 days. Numerous laboratories have failed to confirm that daily injections of PYY(3-36) decrease body weight. Continuous subcutaneous administration of PYY(3-36) by osmotic minipump has been reported to reduce daily food intake in rodents but only during the first 3-4 days of administration. Here we show the effects of different daily patterns of intravenous infusion of PYY(3-36) on food intake, body weight, and adiposity in rats tethered via infusion swivels to computer-controlled pumps. Measurement of food bowl weight recorded by computer every 20 s permitted daily assessment of the instantaneous effects of PYY(3-36) administration on food intake and meal patterns. One-hour intravenous infusions of PYY(3-36) at 30 pmol x kg(-1) x min(-1) every other hour for 10 days produced a sustained reduction in daily food intake of approximately 20% and decreased body weight and adiposity by 7 and 35%, respectively. Thus dosage pattern is critical for producing a sustained effect of PYY(3-36) on food intake and adiposity.


Subject(s)
Adiposity/drug effects , Feeding Behavior/drug effects , Peptide YY/administration & dosage , Peptide YY/pharmacology , Animals , Drug Administration Schedule , Injections, Intravenous , Male , Peptide Fragments , Rats , Rats, Sprague-Dawley , Weight Gain/drug effects
15.
Chem Res Toxicol ; 18(5): 817-24, 2005 May.
Article in English | MEDLINE | ID: mdl-15892575

ABSTRACT

Cigarette smoke is a risk factor for the development of several diseases, but the exact mechanism responsible has not been well-characterized. Because modification, or adducting, of biomolecules is thought to mediate the toxic effects observed from exposure to a wide variety of harmful chemicals, this study investigated the ability of cigarette smoke to produce specific adducts on a peptide to gain insight into the likely effect on cellular proteins. We describe the modification of the epsilon-amino group of lysine contained in a test peptide with stable fluorescent adducts derived from monofunctional aldehydes occurring in cigarette smoke and malonaldehyde, a product of lipid peroxidation. Utilizing high-performance liquid chromatography, fluorescent measurements, and matrix-assisted laser desorption/ionization time-of-flight mass spectroscopy, the 1,4-dihydropyridine-3,5-dicarbaldehyde and 4-methyl-1,4-dihydropyridine-3,5-dicarbaldehyde derivatives of lysine were identified as products of exposure to cigarette smoke extract and malonaldehyde. These data suggest that cigarette smoke may promote the modification of proteins, like those associated with oxidized low-density lipoprotein, and may contribute to smoking-related disease.


Subject(s)
Aldehydes/chemistry , Lipid Peroxidation , Lysine/metabolism , Malondialdehyde/chemistry , Nicotiana/toxicity , Chromatography, High Pressure Liquid , Dihydropyridines/isolation & purification , Fluorescence , Malondialdehyde/metabolism , Proteins/chemistry , Proteins/metabolism , Smoking/adverse effects , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors , Nicotiana/chemistry , Tobacco Smoke Pollution
16.
Am J Physiol Regul Integr Comp Physiol ; 288(6): R1695-706, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15718384

ABSTRACT

Glucagon-like peptide-1(7-36)-amide (GLP-1) is postulated to act as a hormonal signal from gut to brain to inhibit food intake and gastric emptying. A mixed-nutrient meal produces a 2 to 3-h increase in plasma GLP-1. We determined the effects of intravenous infusions of GLP-1 on food intake, sham feeding, and gastric emptying in rats to assess whether GLP-1 inhibits food intake, in part, by slowing gastric emptying. A 3-h intravenous infusion of GLP-1 (0.5-170 pmol.kg(-1).min(-1)) at dark onset dose-dependently inhibited food intake in rats that were normally fed with a potency (mean effective dose) and efficacy (maximal % inhibition) of 23 pmol.kg(-1).min(-1) and 82%, respectively. Similar total doses of GLP-1 administered over a 15-min period were less potent and effective. In gastric emptying experiments, GLP-1 (1.7-50 pmol.kg(-1).min(-1)) dose-dependently inhibited gastric emptying of saline and ingested chow with potencies of 18 and 6 pmol.kg(-1).min(-1) and maximal inhibitions of 74 and 83%, respectively. In sham-feeding experiments, GLP-1 (5-50 pmol.kg(-1).min(-1)) dose-dependently reduced 15% aqueous sucrose intake in a similar manner when gastric cannulas were closed (real feeding) and open (sham feeding). These results demonstrate that intravenous infusions of GLP-1 dose-dependently inhibit food intake, sham feeding, and gastric emptying with a similar potency and efficacy. Thus GLP-1 may inhibit food intake in part by reducing gastric emptying, yet can also inhibit food intake independently of its action to reduce gastric emptying. It remains to be determined whether intravenous doses of GLP-1 that reproduce postprandial increases in plasma GLP-1 are sufficient to inhibit food intake and gastric emptying.


Subject(s)
Appetite Depressants , Eating/drug effects , Gastric Emptying/drug effects , Glucagon/pharmacology , Peptide Fragments/pharmacology , Protein Precursors/pharmacology , Animals , Diet , Dose-Response Relationship, Drug , Glucagon/administration & dosage , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Infusions, Intravenous , Male , Peptide Fragments/administration & dosage , Protein Precursors/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Glucagon/drug effects
17.
Endocrinology ; 146(2): 879-88, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15539554

ABSTRACT

Peptide YY (3-36) [PYY (3-36)] is postulated to act as a hormonal signal from the gut to the brain to inhibit food intake and gastric emptying. A mixed-nutrient meal produces a prolonged 2-3 h increase in plasma levels of both PYY (3-36) and PYY (1-36). We determined the dose-dependent effects of 3-h iv infusions of PYY (3-36) and PYY (1-36) (0.5-50 pmol.kg(-1).min(-1)) at dark onset on food intake in non-food-deprived rats. PYY (3-36) dose-dependently inhibited food intake: the minimal effective dose was 5 pmol.kg(-1).min(-1); the estimated potency (mean effective dose) and efficacy (maximal percent inhibition) were 15 pmol.kg(-1).min(-1) (2.6 nmol/kg) and 47%, respectively. PYY (1-36) was an order of magnitude less potent than PYY (3-36). Similar total doses of PYY (3-36) (0.9-30 nmol/kg) infused during the 15-min period just before dark onset also dose-dependently inhibited food intake, albeit with a lower potency and efficacy. Other experiments showed that PYY (3-36) inhibited food intake in sham-feeding rats and was more effective in reducing intake of a mixed-nutrient liquid diet than 15% aqueous sucrose. We conclude that: 1) iv infusions of PYY (3-36), which are more likely than ip injections to mimic postprandial increases in plasma PYY (3-36), potently inhibit food intake in a dose-dependent manner; 2) PYY (1-36) is an order of magnitude less potent than PYY (3-36); and 3) PYY (3-36) can inhibit food intake independently of its action to inhibit gastric emptying. It remains to be determined whether iv doses of PYY (3-36) that reproduce postprandial increases in plasma PYY (3-36) are sufficient to inhibit food intake.


Subject(s)
Eating/drug effects , Peptide YY/pharmacology , Animals , Dietary Sucrose/pharmacology , Dose-Response Relationship, Drug , Enteral Nutrition , Infusions, Intravenous , Male , Peptide Fragments , Rats , Rats, Sprague-Dawley
18.
Am J Physiol Regul Integr Comp Physiol ; 287(5): R1064-70, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15242829

ABSTRACT

We compared the effects of the two molecular forms of the brain-gut peptide YY (PYY), PYY(1-36) and PYY(3-36), on gastric emptying. Unanesthetized rats received 20-min intravenous infusions of rat PYY(1-36) (0, 1.7, 5, 17, 50, 100, 170 pmol x kg(-1) x min(-1)) and rat PYY(3-36) (0, 0.5, 1.7, 5, 17, 50, 100, 170 pmol x kg(-1) x min(-1)), either alone or combined, and gastric emptying of saline was measured during the last 10 min of infusion. For comparison, human PYY(3-36) was administered at 0, 17, and 50 pmol x kg(-1) x min(-1). Gastric emptying was decreased by 11, 24, 26 and 38% in response to 17, 50, 100, and 170 pmol x kg(-1) x min(-1) of rat PYY(1-36); by 10, 26, 41, 53, and 57% in response to 5, 17, 50, 100, and 170 pmol x kg(-1) x min(-1) of rat PYY(3-36); and by 35 and 53% in response to 17 and 50 pmol x kg(-1) x min(-1) of human PYY(3-36), respectively. Estimated ED50s were 470 and 37 pmol x kg(-1) x min(-1) for rat PYY(1-36) and PYY(3-36), respectively. In general, within an experiment, coadministration of PYY(1-36) and PYY(3-36) inhibited gastric emptying by an amount that was comparable to that produced when either peptide was given alone. We conclude that 1) intravenous infusion of PYY(1-36) and PYY(3-36) each produces a dose-dependent inhibition of gastric emptying in rats, 2) PYY(3-36) is an order of magnitude more potent than PYY(1-36) in inhibiting gastric emptying, 3) human PYY(3-36) and rat PYY(3-36) inhibit gastric emptying similarly, and 4) PYY(1-36) and PYY(3-36) do not appear to interact in an additive or synergistic manner to inhibit gastric emptying.


Subject(s)
Gastric Emptying/drug effects , Peptide YY/pharmacology , Pharmacology , Animals , Dose-Response Relationship, Drug , Humans , Infusions, Intravenous , Male , Peptide Fragments , Rats , Rats, Sprague-Dawley
19.
Am J Physiol Regul Integr Comp Physiol ; 287(3): R568-74, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15130879

ABSTRACT

Amylin is postulated to act as a hormonal signal from the pancreas to the brain to inhibit food intake and regulate energy reserves. Amylin potently reduces food intake, body weight, and adiposity when administered systemically or into the brain. Whether selective blockade of endogenous amylin action increases food intake and adiposity remains to be clearly established. In the present study, the amylin receptor antagonist acetyl-[Asn(30), Tyr(32)] sCT-(8-32) (AC187) was used to assess whether action of endogenous amylin is essential for normal satiation to occur. Non-food-deprived rats received a 3- to 4-h intravenous infusion of AC187 (60-2,000 pmol.kg(-1).min(-1)), either alone or coadministered with a 3-h intravenous infusion of amylin (2.5 or 5 pmol.kg(-1).min(-1)) or a 2-h intragastric infusion of an elemental liquid diet (4 kcal/h). Infusions began just before dark onset. Food intake and meal patterns during the first 4 h of the dark period were determined from continuous computer recordings of changes in food bowl weight. Amylin inhibited food intake by approximately 50%, and AC187 attenuated this response by approximately 50%. AC187 dose-dependently stimulated food intake (maximal increases from 76 to 171%), whether administered alone or with an intragastric infusion of liquid diet. Amylin reduced mean meal size and meal frequency, AC187 attenuated these responses, and AC187 administration alone increased mean meal size and meal frequency. These results support the hypothesis that endogenous amylin plays an essential role in reducing meal size and increasing the postmeal interval of satiety.


Subject(s)
Amyloid/physiology , Eating/physiology , Receptors, Peptide/physiology , Amyloid/antagonists & inhibitors , Animals , Anorexia/chemically induced , Anorexia/physiopathology , Eating/drug effects , Enteral Nutrition , Food, Formulated , Injections, Intravenous , Intubation, Gastrointestinal , Islet Amyloid Polypeptide , Peptide Fragments , Peptides/administration & dosage , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Islet Amyloid Polypeptide , Receptors, Peptide/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...