Neuropeptide Y (NPY) is thought to play a key role in stimulating feeding, thus making NPY receptors attractive appetite suppressant drug targets for treating obesity. Because the orexigenic effects of NPY have been ascribed to actions at the NPY Y5 receptor, we have determined the role of this receptor in feeding in rats, using a small molecule antagonist of this receptor. NPY5RA-972 is a selective and potent (<10 nmol/l) NPY Y5 receptor antagonist. This compound is central nervous system (CNS) penetrant, and an oral dose of 10 mg/kg NPY5RA-972 to rats produced concentrations in cerebrospinal fluid that Xgreatly exceeded the in vitro IC50 (inhibitory concentration 50%). Indeed, at doses to rats as low as 1 mg/kg, NPY5RA-972 inhibited feeding induced by intracerebroventricular (ICV) administration of a selective NPY Y5 agonist ([cPP1–7,NPY19–23,Ala31,Aib32,Gln34]-hPP). However, in the dose range 1–10 mg/kg, NPY5RA-972 had no significant effect on food intake in Wistar rats induced to feed by either ICV NPY or 24 h fasting or in free-feeding Wistar or obese Zucker rats. Chronic administration of NPY5RA-972 (10 mg/kg twice daily) had no effect on food intake or body weight in either free-feeding Wistar rats or dietary obese rats. These data indicate that NPY5RA-972 is a potent, selective, orally active, and CNS-penetrant antagonist of the NPY Y5 receptor that prevents feeding driven by activation of this receptor. The data obtained with this antagonist indicate that the NPY Y5 receptor is not a major regulator of feeding in the rat.

Research into appetite control during the past decade has been ignited by the discovery of leptin and fueled by the recognition of obesity as a widespread and rapidly growing disease (1). Leptin is a fat-derived hormone that signals energy (fat) storage levels to the hypothalamus. A number of previously identified (e.g., neuropeptide Y [NPY], melanin-concentrating hormone, corticotropin-releasing factor, galanin, neuromedin U, bombesin and pro-opiomelanocortin peptides) and newly discovered (agouti-related protein, cocaine and amphetamine-regulated transcript, urocortin, orexin, ghrelin) neuropeptides have become recognized as central nervous system (CNS) targets of leptin action (25). Many of these peptides are known to influence feeding in experimental species, and their receptors have been proposed as possible targets for appetite suppressant drugs. Corroborative evidence supporting these hypotheses from human (6,7) and rodent mutations (810) is limited, but compensation and redundancy have been invoked as explanations for unexpected negative findings (11). Ultimately, the generation of potent and selective modulators of these peptide pathways is key to our ability to explore the biology of appetite control and to defining which of these pathways are likely to be the best targets for appetite suppressant drugs.

NPY is a 36–amino acid peptide that is probably the most studied putative neuropeptide regulator of appetite. In rodents, NPY is expressed in regions of the hypothalamus thought to be important in the regulation of feeding. Its level of expression is sensitive to energy status, and its administration stimulates feeding and reduces energy expenditure; immunoneutralization of endogenous NPY inhibits feeding (reviewed in 12,13). Collectively, these types of data have resulted in the recognition of NPY as a key regulator of appetite even though targeted gene deletion of NPY provides only limited evidence to support this concept (14,15).

The effects of NPY are mediated by distinct receptor subtypes; NPY Y1 and NPY Y5 have become recognized as the most likely candidates for the mediation of the effects of NPY on food intake (12,13, 16). In particular, the importance of NPY Y5 has been suggested by the parallel pharmacology of the receptor in vitro and feeding in rodents (1724), and the inhibitory effects of NPY Y5 receptor antisense oligonucleotides (2528), NPY Y5 deficiency (29), and an NPY Y5 antagonist (30) on NPY-induced feeding in rodents. We investigated the role the NPY Y5 receptor in feeding in the rat using a previously described peptidic NPY Y5 selective agonist [cPP1–7, NPY19–23,Ala31,Aib32,Gln34]-hPP (20) and a novel, small molecular weight, potent, and selective NPY Y5 antagonist (9-isopropyl-4-methyl-3-(4-morpholinecarbonylamino)-9H-carbazole, termed NPY5RA-972) (31). The data described herein argue that, contrary to many previous data, NPY Y5 is not a significant regulator of feeding behavior in the rat.

Reagents and materials.

NPY, human PYY, [cPP1–7,NPY19–23, Ala31,Aib32,Gln34]-hPP, and sibutramine were purchased from Bachem (Saffron Walden, Essex, U.K.), Sigma Aldrich, Tocris Cookson (Bristol, U.K.), and St. Andrews ChemTech (Fife, U.K.), respectively. NPY5RA-972 (31) was synthesized within the Medicinal Chemistry laboratories at AstraZeneca (Alderley Park, U.K.).

Membrane binding assays.

Hi5 insect cells transiently (48 h) transfected (baculovirus-infected) with either rat or human NPY Y5 receptor were used for preparation of NPY Y5 membranes. Human NPY Y1, NPY Y2, and NPY Y4 membranes were prepared from SK-N-MC, KAN-TS, and stable NPY Y4 expressing CHO cells, respectively. Membranes were prepared by sonication (3 × 15 s) in ice-cold hypotonic buffer (12.5 mmol/l Tris, 1.25 mmol/l EDTA, 2.5% sucrose [pH 7.4]) containing complete protease inhibitor tablets (1/100 ml; Roche Molecular Biochemicals). The lysate was layered onto a 41% sucrose cushion and centrifuged at 100,000g for 1 h. The membrane layer was harvested and stored (in 50 mmol/l Tris, 5 mmol/l EDTA, 10% sucrose [pH 7.4]) at −80°C until use.

Binding assays were performed in presiliconized, round-bottomed, polypropylene 96-well plates (Corning Costar). Compounds were dissolved in DMSO and diluted (20-fold) in binding buffer (50 mmol/l HEPES, 2.5 mmol/l CaCl2, 1 mmol/l MgCl2, 0.5% BSA [pH 7.4]). Each incubate had 0.01 μCi of [125I]porcine peptide YY (Amersham Pharmacia Biotech) added to 10–80 μl of membranes (sufficient to give a specific binding of 1,500 cpm) and 10 μl of compound solution. After a 2-h incubation at room temperature, incubates were filtered onto Canberra Packard GF/C 96-well filter plates (Packard Instruments) pretreated with 0.5% polyethylenimine using a Brandel harvester. The plates were washed twice with wash buffer (binding buffer + 0.5 mmol/l NaCl). The filter plates were dried overnight and counted on a Canberra Packard Top Count (Packard Instruments) after addition of 20 μl of Microscint 40 (Packard Instruments) to each well.

Reporter gene functional assay.

Recombinant HEK293 cells stably expressing the rat NPY Y5 receptor and also a reporter cassette consisting of a cyclic AMP driven β-galactosidase gene were used. The cells were grown in T175 flasks to 80–90% confluence in Dulbecco’s modified Eagle’s medium (DMEM) plus 10% FCS, 1% penicillin/streptomycin, and 1% l-glutamine. After harvesting, cells were washed and resuspended in indicator-free DMEM. Cells (2 × 104 cells in 50 μl) were preincubated (in triplicate) with compounds (in 20 μl) for 5 min at room temperature in flat-bottomed polystyrene 96-well plates (Corning Costar). NPY (20 μl) was added to each of the wells for a 5-min incubation at room temperature. Ten microliters of 10 μmol/l forskolin (Sigma Aldrich) was added to start the incubation of 3.75 h at 37°C. After incubation, the plates were cooled at room temperature before the addition of 50 μl of the chlorophenol red-β -d-galacto-pyranoside cocktail (CPRG) (0.6% SDS, 1.5 mmol/l CPRG (Roche Molecular Biochemicals), 24 mmol/l Na2HPO4(2H2O), 40 mmol/l NaH2PO4(2H2O), 10 mmol/l KCl, and 1 mmol/l MgSO4(7H2O). After an overnight incubation at room temperature, the absorbance of each plate was read on a Tecan SpectraFluor at 570 nm.

Animals.

Male Wistar and obese (fa/fa) Zucker rats (ZuckerfLIS/Alpkh) were obtained from the animal breeding unit at AstraZeneca. Sprague-Dawley rats were obtained from Charles River (Kent, U.K.). Unless otherwise stated, animals were housed individually in a temperature- and humidity-controlled environment with a 12-h light:12-h dark cycle (lights on 06:00 h) and provided with standard rat diet (RM1, 2.61 kcal/g; Special Diet Services, Essex, U.K.) and water ad libitum.

Oral gavage.

Suspensions of NPY5RA-972 or sibutramine were prepared in hydroxypropylmethylcellulose (HPMC; 0.5% wt/vol), polysorbate 80 (Tween 80; 0.1% wt/vol) in water. Vehicle or NPY5RA-972 was dosed by oral gavage (75 mm × 16-G curved oral dosing cannula; IMS, Cheshire, U.K.) at a volume of 2 ml/kg.

Intacerebroventicular cannulation and injection.

Male Wistar rats (220–270 g) were anesthetized with Halothane and secured in a stereotaxic apparatus (Stoelting, Wood Dale, IL). With the incisor bar set at −3.3 mm below the intra-aural line, a guide cannula (8-mm length; Plastics One, Roanoke, VA) was implanted in the third ventricle at the following coordinates, according to the rat brain atlas of Paxinos and Watson (32): lateral 0 mm; anterior-posterior −3 mm; dorsoventral −8 mm. Cannulae were secured with two screws and dental compomer (Dyract AP, Dentsply, Wright Cottrell, Manchester, U.K.) cured with an ultraviolet light (QHL75, Dentsply). After suturing, a dummy needle was inserted to keep the cannula patent, and rats were allowed to recover from anesthesia. Rats were left to recover for 10–14 days, and all animals used in subsequent experiments had recovered at least to presurgical weight by the day of experimentation.

Peptides were dissolved in sterile water and administered intracerebroventricularly via an injection cannula (projecting 1 mm below the tip of the guide cannula) attached to a length of siliconized tubing (0.51 mm internal diameter) connected to a 25-μl Hamilton microsyringe. Intracerebroventricular (ICV) injections (5 μl) were administered to conscious, unrestrained rats over a period of 60 s.

Pharmacokinetic profile of NPY5RA-972.

Male Wistar rats (n = 3) were dosed by mouth (p.o.) with 30 μmol/kg NPY5RA-972, and plasma was obtained from 9–10 whole-blood samples withdrawn over a 24-h period (total volume of blood ∼2 ml, replaced by an equal volume of saline) after dosing. NPY5RA-972 concentrations in plasma were determined by high-performance liquid chromatography-ultraviolet (285 nm) analysis.

In separate experiments, male Wistar rats (350–400 g) were dosed with NPY5RA-972 (10–80 mg/kg p.o.). One hour later, they were terminally anesthetized with sagatal (pentobarbitone, 60 mg/ml, 0.6 ml/rat) and mounted in a stereotaxic apparatus, and a cerebrospinal fluid sample (∼100 μl) was collected using a 25-G butterfly needle inserted through the muscles of the neck and piercing the dura overlying the cisterna magna. Cerebrospinal fluid (CSF) samples that showed any discoloration were discarded; only clear samples were subject to analysis. CSF and plasma samples collected terminally were processed for subsequent analysis of compound concentrations.

Effects of NPY and [cPP1–7,NPY19–23,Ala31,Aib32,Gln34]-hPP on food intake.

Either NPY or [cPP1–7,NPY19–23, Ala31,Aib32,Gln34]-hPP was injected intracerebroventricularly into male Wistar rats (n = 5–7) in the free-feeding state during the early light phase (commencing 08:00–10:00 h). Animals were returned to their home cage that contained a preweighed amount of food, and the food that remained at 1, 2, 4, and 6 h was determined on a Mettler Toledo PG2002-S balance (Fischer Scientific, Leicestershire, U.K.), corrected for spillage, and recorded to the nearest 0.1 g.

Effects of NPY5RA-972 on food intake induced by NPY or [cPP1–7,NPY19–23,Ala31,Aib32,Gln34]-hPP.

NPY5RA-972 (1–10 mg/kg) or vehicle was dosed (p.o.) to male Wistar rats (n = 4–7) in the free-feeding state during the early light phase (commencing 08:00–10:00 h). One hour later, these rats received an ICV injection of either [cPP1–7,NPY19–23,Ala31,Aib32,Gln34]-hPP (0.2 or 0.6 nmol/rat) or NPY (0.2 or 2.0 nmol/rat). Cumulative food intake was measured as described.

Acute effects of NPY5RA-972 on food intake in fasted, free-feeding, and obese Zucker rats.

Male Wistar rats (219–267 g, n = 8–12) were fasted from 10:00 h and 23 h later dosed (p.o.) with either vehicle or NPY5RA-972 (1–10 mg/kg). One hour later, preweighed food hoppers were returned to the animals, and cumulative food intake was measured 1, 2, 4, and 6 h later.

Free-feeding male Wistar rats (240–306 g, n = 8 per group) or male obese Zucker rats (472–492 g, n = 7–12) were dosed p.o. with either vehicle or NPY5RA-972 (1–10 mg/kg) 1 h before the onset of the dark-phase (18:00 h) and cumulative food intake measured from 18:00 h for either 2, 4, 15, and 24 h or 6, 14, and 24 h, respectively.

Chronic effects of NPY5RA-972 and sibutramine on food intake and body weight in normal and dietary obese rats.

To ascertain a suitable twice-daily dosing regimen for chronic studies that provided 24-h blockade of NPY Y5 receptors in brain, we determined the effects of 12-h predosing of NPY5RA-972 on food intake induced by [cPP1–7,NPY19–23,Ala31, Aib32,Gln34]-hPP. Male Wistar rats (n = 6/group) were dosed with NPY5RA-972 (1–10 mg/kg) at 21:00 h and received an ICV injection of [cPP1–7,NPY19–23,Ala31,Aib32,Gln34]-hPP (0.6 nmol/rat) 12 h later. Food intake for the 2 h after [cPP1–7,NPY19–23,Ala31,Aib32, Gln34]-hPP injection was determined as described above. As a positive control, an additional group of rats were administered NPY5RA-972 (3 mg/kg p.o., n = 5) 1 h before [cPP1–7, NPY19–23,Ala31,Aib32, Gln34]-hPP.

In chronic studies that used previously unmanipulated Wistar rats, NPY5RA-972 (10 mg/kg, n = 7) or vehicle (n = 6) was dosed (p.o.) twice daily (at 06:00 h and 16:00 h). For 3 days before commencement of dosing with compound, rats were accustomed to handling and dosing with vehicle only. Food intake and body weight were measured daily. A positive control group (n = 7) receiving sibutramine was included in the chronic studies. Sibutramine is a marketed appetite-suppressant drug that acts principally by inhibiting the reuptake of serotonin and norepinephrine (33). Sibutramine was dosed at 3 mg/kg once daily (at 16:00 h), a dose that was previously proved to be effective in our and others’ hands (34), and is the maximum dose we have found that avoids marked behavioral disruption. Sibutramine-treated rats also received a dose of vehicle (at 06:00 h) to maintain consistency of procedures across all groups.

In diet-induced obese studies, male Wistar rats were fed a high-energy diet (D12266B, 4.41 kcal/g; Research Diets). This rodent diet is designed to distinguish obesity-prone and obesity-resistant phenotypes and is similar to that used by Levin and colleagues (35,36) in their studies of susceptibility and resistance to obesity. Energy intake in rats fed a high-energy diet is expressed as kcal/d.

Data presentation and statistical analysis.

Unless otherwise stated, results are presented as mean ± SE. In vitro dose-response curves were calculated and plotted using a nonlinear least squares curve-fitting routine within Origin 5 (Microcal Software). All animal experiments were based on a between-individual design with each animal being tested on a single occasion in a single experiment. Feeding and body weight data were analyzed by one-way ANOVA followed by Bonferroni multiple comparison test. A two-tailed probability of P < 0.05 was considered statistically significant.

In vitro profile of NPY5RA-972 and [cPP1–7,NPY19–23,Ala31,Aib32,Gln34]-hPP.

NPY5RA-972 bound with high affinity to both the human and the rat Y5 receptor (half-maximal inhibitory concentration [IC50], 3.1 ± 1.0 nmol/l and 9.3 ± 1.2 nmol/l, respectively; Table 1). In contrast, 10 μmol/l NPY5RA-972 displayed no binding to human NPY Y1, NPY Y2, or NPY Y4 receptors and showed at least 1,000-fold selectivity for NPY Y5 in a commercially available panel (MDS Pharma Services) of 129 different bindings assays (including assays for NPY, muscarinic, serotonergic opioid, melanocortin, galanin, cholecystokinin, cannabinoid, neurotensin, glucagon-like peptide receptors, and serotonin transporter).

In a functional reporter assay for NPY Y5 antagonism, NPY5RA-972 completely reversed the suppressive effect of 10 nmol/l NPY on 1 μmol/l forskolin-stimulated HEK 293 cells stably expressing the rat Y5 receptor (Table 1). NPY5RA-972 displayed no agonist activity as indicated by a lack of effect of 10 μmol/l of the compound in the absence of NPY in this reporter assay (data not shown).

[cPP1–7,NPY19–23,Ala31,Aib32,Gln34]-hPP is an NPY Y5 selective agonist recently described by Cabrele et al. (20). In broad agreement with Cabrele et al. (20), we found that [cPP1–7,NPY19–23,Ala31,Aib32,Gln34]-hPP binds with high affinity to rat and human NPY Y5, has potent agonist activity in the rat NPY Y5 reporter assay, and has a high degree of selectivity for the NPY Y5 receptor (>5,000-fold versus NPY Y1, ∼ 200-fold versus NPY Y2, ∼500-fold versus NPY Y4; Table 1).

Pharmacokinetics of NPY5RA-972 in plasma and CSF.

The mean (of three rats) time to maximum plasma concentration (Tmax) after oral gavage of 30 μmol/kg (=10.5 mg/kg) NPY5RA-972 was 1 h, at which time the total concentration (Cmax) was ∼35 μmol/l. At a measured in vitro protein binding of 99.1%, predicted free plasma concentrations of NPY5RA-972 at Tmax was 315 nmol/l. The half-life of NPY5RA-972 after oral dosing was 6.4 h, the 24-h area under the curve was ∼ 350 μmol · l−1 · h−1, and even at 24 h after dosing total plasma concentrations were still >3 μ mol/l.

In separate dose-response studies (10–80 mg/kg p.o.), total plasma concentrations of NPY5RA-972 at 1 h increased with dose but did not scale linearly. CSF concentrations of NPY5RA-972 (153 ± 35 nmol/l) after a dose of 10 mg/kg (p.o.) massively exceeded the IC50 of NPY5RA-972 (by 16- to 50-fold; Tables 1 and 2), with higher doses producing even greater CSF concentrations.

Effects of NPY and [cPP1–7,NPY19–23,Ala31,Aib32, Gln34]-hPP on food intake.

[cPP1–7,NPY19–23,Ala31, Aib32,Gln34]-hPP injected intracerebroventricularly produced a marked and dose-dependent increase in food intake (Fig. 1A). The maximum dose of this NPY Y5 selective agonist seemed slightly lower (0.6 nmol) than that for NPY (2.0 nmol; Fig. 1A and B). The time course of effect of these two peptides seemed slightly different, with the NPY Y5 selective agonist having a less pronounced initial but more sustained effect (Fig. 1C), a profile identical to what we have observed with the less selective NPY Y5 agonist hPP (data not shown).

Effects of NPY5RA-972 on food intake induced by NPY or [cPP1–7,NPY19–23,Ala31,Aib32,Gln34]-hPP.

NPY5RA-972 (p.o.) inhibited food intake in rats induced by [cPP1–7,NPY19–23,Ala31, Aib32,Gln34]-hPP (ICV). NPY5RA-972 (10 mg/kg) completely abolished food intake induced by either submaximum (0.2 nmol) or maximum (0.6 nmol) doses of the NPY Y5 agonist (Fig. 2A). Even at doses as low as 1 mg/kg, NPY5RA-972 produced a marked and significant inhibition of the feeding response to NPY Y5 agonist (Fig. 2B).

In contrast to the abolition of the feeding response to a maximum dose of NPY Y5 selective agonist, NPY5RA-972 (3 mg/kg p.o.) had no effect on food intake induced by a maximum dose (2.0 nmol) of NPY infused 1 h later (Fig. 3A). Furthermore, even at higher doses of NPY5RA-972 (10 mg/kg), this antagonist had no effect on feeding induced by a submaximum dose (0.2 nmol) of NPY (Fig. 3B).

Acute effects of NPY5RA-972 on food intake in fasted, free-feeding, and obese Zucker rats.

NPY5RA-972 (1–10 mg/kg p.o.) dosed 1 h before presentation of food had no significant effect on food intake for the ensuing 6 h (Fig. 4A). NPY5RA-972 (1–10 mg/kg) also failed to affect the fast-induced feeding response in another strain of rat (Sprague-Dawley, data not shown). Similarly, in free-feeding Wistar rats (Fig. 4B) or obese Zucker rats (Fig. 4C) dosed 1 h before the onset of the dark cycle (main feeding phase), NPY5RA-972 had no significant effect on food intake.

Chronic effects of NPY5RA-972 and sibutramine on food intake and body weight in normal and dietary obese rats.

At a dose of 10 mg/kg, NPY5RA-972 (but not lower doses) 12 h predosing markedly inhibited food intake induced by the NPY Y5 selective agonist for 2 h (Fig. 5), indicating that this dose is sufficient to antagonize NPY Y5 receptors in the brain for at least 12–14 h. Therefore, a dosing regimen of 10 mg/kg twice daily was selected for chronic studies.

In normal Wistar rats, 3 mg/kg sibutramine produced a marked (∼ 30%) inhibition of food intake on the first day of dosing (Fig. 6A). Consistent with published data, the effects of sibutramine on food intake diminished with time (34,37), although cumulative food intake over the 9-day study was significantly (P < 0.001) lower in sibutramine-treated (213.3 ± 5.7 g) than in vehicle-treated (260.2 ± 3.0 g) rats. Sibutramine also significantly reduced overall body weight gain (vehicle 30 ± 2 g, sibutramine 14 ± 3 g; P < 0.001; Fig. 6B). However, chronic antagonism of NPY Y5 with NPY5RA-972 produced no significant effect on either food intake (cumulative food intake 252.3 ± 6.3 g; Fig. 6A) or body weight gain (27 ± 1g; Fig. 6B) compared with vehicle-treated controls.

Five-week-old Wistar rats were placed on a highly palatable high-energy diet or maintained on normal rat diet for a period of 12 weeks. After 10 weeks on the high-energy diet, only rats that exhibited clear hyperphagia and obesity compared with rats fed a normal diet were selected. Indeed, the calorie intakes (107 ± 4 vs. 72 ± 3 kcal/d) and body weights (596 ± 8g vs. 478 ± 18 g) were markedly higher in this selected obese cohort compared with the age-matched controls that were fed the regular diet. This diet-induced obese (DIO) cohort was randomly divided into three groups that were dosed (p.o.) with vehicle (twice daily), sibutramine (3 mg/kg, once daily) or NPY5RA-972 (10 mg/kg, twice daily). As in the previous experiment, sibutramine produced a significant (P < 0.001) inhibition of energy intake (cumulative energy intake 791 ± 26 kcal) compared with vehicle-treated (1,227 ± 55 kcal) rats, although its effects in DIO animals seemed far greater (Fig. 7A) than in Wistar rats that were fed a regular diet (Fig. 6A). Indeed, sibutramine not only slowed weight gain but also caused significant (P < 0.001) weight loss in DIO rats over the 12-day study period (vehicle 14.0 ± 6.1 g, sibutramine −37.0 ± 7.8 g; Fig. 7B). In contrast, NPY5RA-972 had no significant effect on either calorie intake (cumulative intake 1,199 ± 87 kcal) or body weight (12 ± 9g; Fig. 7).

A number of lines of evidence have suggested that NPY Y5 is a major receptor subtype that mediates the effects of NPY on feeding and has attracted much academic and industrial interest as a possible appetite suppression approach to the treatment of obesity (12, 30,38). Notably, the majority of these data have been generated in rodents. Supportive data in humans has been restricted to a neuroanatomical distribution similar to rodents (39,40) and to preliminary human genetic evidence in a very specific cohort (41). Indeed, other human genetic studies have resulted in negative findings (42,43). The data reported herein identify NPY5RA-972 as a potent and selective NPY Y5 receptor antagonist with good systemic and CNS exposure after oral dosing in rats, suggesting that it is a useful tool with which to explore the role(s) of NPY Y5 in physiological processes. Coadministration studies indicated that NPY5RA-972 prevented feeding induced by the NPY Y5 selective agonist [cPP1–7,NPY19–23, Ala31, Aib32,Gln34]-hPP. The most obvious conclusion of these experiments is that NPY5RA-972 after oral dosing occupies sufficient NPY Y5 receptors in the brain to prevent NPY Y5 agonist-induced feeding. The possibility that this apparent agonism-antagonism reflects activities at alternative, non-NPY Y5 feeding-related targets cannot be totally disproved. However, 1) [cPP1–7,NPY19–23,Ala31,Aib32,Gln34]-hPP is highly selective for NPY Y5 compared with other known NPY receptors (20 and present study); 2) NPY5RA-972 shows at least 1,000-fold selectivity against a panel of 129 receptors, enzymes, and transporters that included a number of receptors relevant to feeding; and 3) [cPP1–7,NPY19–23, Ala31,Aib32,Gln34]-hPP and NPY5RA-972 are of different structural classes (peptidic versus nonpeptidic), optimized for NPY Y5 potency and selectivity by two different groups using completely different chemical strategies, making it highly unlikely that both were unwittingly optimized for activity at alternative identical targets. It seems much more likely that their activities in co-administration experiments reflect agonism and antagonism at the NPY Y5 receptor, leading us to conclude that at the doses used in this study, NPY5RA-972 is an ideal tool to test hypotheses regarding the role of the NPY Y5 receptor in the rat.

Although low doses (1–10 mg/kg p.o.) of NPY5RA-972 inhibited food intake elicited by a selective NPY Y5 receptor agonist, it had no effect on feeding as a result of NPY (ICV), fasting, free-feeding in normal, genetically obese, or dietary obese rats. These findings strongly suggest that the NPY Y5 receptor is not a major physiological regulator of feeding in rats.

A previous study with a different NPY Y5 antagonist, CGP 71683A (30), drew very different conclusions regarding the role of NPY Y5 in feeding in the rat. CGP 71683A produces marked inhibitory effects in a variety of rat feeding models, including the ICV administration of NPY. NPY5RA-972 belongs to a series of carbazole compounds, and indeed within this series we found potent in vitro NPY Y5 antagonists that have impressive hypophagic activity in vivo in a variety of rodent feeding models (data not shown). However, we found that an exemplary compound with impressive hypophagic activity was also effective at inhibiting food intake in NPY Y5-deficient mice (data not shown), indicating that NPY Y5 antagonism was not its primary mode of action. Likewise, it now seems that CGP 71683A has been discredited as an in vivo tool to explore the biology of NPY Y5 as a result of its activity at serotonergic transporters and muscarinic receptors (44) and its efficacy in NPY-deficient (45) and NPY Y5-deficient mice (46). Clearly, it cannot be assumed that demonstrated in vitro pharmacology and in vivo hypophagic effects share a “cause-effect relationship.” In this respect, the utility of receptor knockout mice to define the molecular mode of action of drugs cannot be overemphasized. The mechanism of action by which other NPY Y5 antagonists (47,48) reported to produce marked hypophagic effects in rodents is unknown, but notably no data have been presented to indicate that antagonism of NPY Y5 is their primary mode of action. We believe that the present data show convincingly that NPY5RA-972 potently antagonizes NPY Y5 receptors in the brain but fails to affect feeding in a variety of rat feeding models. Emerging preliminary data with a range of other small molecular weight NPY Y5 antagonists (38, 4952) are consistent with our findings.

It seems pertinent to revisit the evidence supporting a role for Y5 in feeding in rodents. This primarily consisted of data showing similarities of the pharmacology of NPY Y5 receptors in vitro and the pharmacology of NPY-related peptides on feeding in vivo (1719,23). Although this conclusion has been disputed (53,54), it is now clear that selective activation of NPY Y5 stimulates feeding in rats (2022). The present study therefore argues that the hyperphagic effect of selective receptor activation is not necessarily a good predictor of the importance of that receptor in feeding under more physiological circumstances. Studies using antisense oligonucleotides to downregulate NPY Y5 expression have also supported the hypothesis that full expression of this receptor is required to elicit the full hyperphagic effects of NPY and that NPY Y5 is important in the regulation of feeding in normal rats (2528). The explanation for the differences in findings with antisense and the effects of an antagonist are unknown, but this discrepancy serves to indicate that such antisense studies are not always predictive of the effects of pharmacological manipulation.

The lack of a hypophagic phenotype of NPY Y5-deficient mice clearly is not supportive of a major role for NPY Y5 in regulating feeding in mice (29). Furthermore, studies indicating that NPY Y5-deficient mice have a reduced hyperphagic response to ICV NPY (29) have now been contradicted (55), and such knockout studies equally indicate that NPY Y1 (or other NPY receptor) is a likely receptor subtype mediating the effects of NPY on food intake. Indeed, a recent study of the effects of a selective NPY Y1 antagonist in NPY-treated wild-type, NPY Y1-deficient, and NPY Y5-deficient mice provides compelling evidence for the involvement of NPY Y1 in NPY-induced feeding (55). A number of other groups have shown that selective NPY Y1 antagonism inhibits NPY-induced feeding in rats (12,13,38, 48).

Little is known about the function of NPY Y5 in rodent brain other than its putative role in feeding and its possible mediation of the antiseizure activity of NPY (56). The development of highly potent and selective agonists and antagonists for NPY Y5 should now permit a more detailed investigation of the biology of this receptor.

FIG. 1.

Effects of NPY Y5 selective agonist [cPP1–7, NPY19–23,Ala31, Aib32, Gln34]-hPP and NPY on food intake. NPY Y5 selective agonist (n = 5–6; A) or NPY (n = 6–7; B) was administered into the third cerebroventricle of male Wistar rats during the light cycle, and food intake was measured 1, 2, 4, and 6 h later. After dose responses were constructed for these two peptides, a direct comparison of maximum doses (NPY Y5 selective agonist 0.6 nmol; NPY 2 nmol) was performed (n = 6 per group; C). ANOVA indicated significant intergroup differences at each time point in all three experiments. *P < 0.05, **P < 0.01, *** P < 0.001 versus control; #P < 0.05, ## P < 0.01 versus 2.0 nmol NPY.

FIG. 1.

Effects of NPY Y5 selective agonist [cPP1–7, NPY19–23,Ala31, Aib32, Gln34]-hPP and NPY on food intake. NPY Y5 selective agonist (n = 5–6; A) or NPY (n = 6–7; B) was administered into the third cerebroventricle of male Wistar rats during the light cycle, and food intake was measured 1, 2, 4, and 6 h later. After dose responses were constructed for these two peptides, a direct comparison of maximum doses (NPY Y5 selective agonist 0.6 nmol; NPY 2 nmol) was performed (n = 6 per group; C). ANOVA indicated significant intergroup differences at each time point in all three experiments. *P < 0.05, **P < 0.01, *** P < 0.001 versus control; #P < 0.05, ## P < 0.01 versus 2.0 nmol NPY.

Close modal
FIG. 2.

Effect of NPY Y5 selective antagonist NPY5RA-972 on food intake induced by the NPY Y5 selective agonist [cPP1–7,NPY19–23, Ala31,Aib32,Gln34]-hPP. A: Vehicle or NPY5RA-972 (10 mg/kg) was dosed by oral gavage (p.o.) 1 h before administration of NPY Y5 selective antagonist (0.2 or 0.6 nmol) into the third cerebroventricle of male rats during the light cycle. ANOVA indicated significant intergroup differences at all time points. **P < 0.01, *** P < 0.001 versus 0.2 nmol NPY Y5 agonist; ###P < 0.001 versus 0.6 nmol NPY Y5 agonist (n = 5–6). B: Vehicle or NPY5RA-972 (1, 3, or 10 mg/kg) was dosed by oral gavage (p.o.) 1 h before administration of 0.6 nmol of NPY Y5 agonist into the third cerebroventricle of male rats during the light cycle. ANOVA indicated significant intergroup differences at the 2-h and 4-h time points. * P < 0.05, **P < 0.01 versus. 0.6 nmol NPY Y5 agonist (n = 4–6).

FIG. 2.

Effect of NPY Y5 selective antagonist NPY5RA-972 on food intake induced by the NPY Y5 selective agonist [cPP1–7,NPY19–23, Ala31,Aib32,Gln34]-hPP. A: Vehicle or NPY5RA-972 (10 mg/kg) was dosed by oral gavage (p.o.) 1 h before administration of NPY Y5 selective antagonist (0.2 or 0.6 nmol) into the third cerebroventricle of male rats during the light cycle. ANOVA indicated significant intergroup differences at all time points. **P < 0.01, *** P < 0.001 versus 0.2 nmol NPY Y5 agonist; ###P < 0.001 versus 0.6 nmol NPY Y5 agonist (n = 5–6). B: Vehicle or NPY5RA-972 (1, 3, or 10 mg/kg) was dosed by oral gavage (p.o.) 1 h before administration of 0.6 nmol of NPY Y5 agonist into the third cerebroventricle of male rats during the light cycle. ANOVA indicated significant intergroup differences at the 2-h and 4-h time points. * P < 0.05, **P < 0.01 versus. 0.6 nmol NPY Y5 agonist (n = 4–6).

Close modal
FIG. 3.

Effect of NPY Y5 selective antagonist NPY5RA-972 on food intake induced by NPY or the NPY Y5 selective agonist [cPP1–7, NPY19–23,Ala31,Aib32, Gln34]-hPP. A: Vehicle or NPY5RA-972 (3 mg/kg) was dosed by oral gavage (p.o.) 1 h before administration of NPY Y5 agonist (0.6 nmol) or NPY (2.0 nmol) into the third cerebroventricle of male rats during the light cycle. Control animals received vehicle (HPMC/Tween, p.o.) followed 1 h later by ICV vehicle (sterile water). ANOVA indicated significant intergroup differences at all time points. *P < 0.05, *** P < 0.001 versus 0.6 nmol NPY Y5 agonist, not significant (ns) versus 2.0 nmol NPY (n = 5–6). B: Vehicle or NPY5RA-972 (10 mg/kg) was dosed by oral gavage (p.o.) 1 h before administration of NPY Y5 agonist (0.6 nmol) or NPY (0.2 nmol) into the third cerebroventricle of male rats during the light cycle. ANOVA indicated significant intergroup differences at all time points. *P < 0.05, *** P < 0.001 versus 0.6 nmol NPY Y5 agonist, not significant (ns) versus 0.2 nmol NPY (n = 6–7).

FIG. 3.

Effect of NPY Y5 selective antagonist NPY5RA-972 on food intake induced by NPY or the NPY Y5 selective agonist [cPP1–7, NPY19–23,Ala31,Aib32, Gln34]-hPP. A: Vehicle or NPY5RA-972 (3 mg/kg) was dosed by oral gavage (p.o.) 1 h before administration of NPY Y5 agonist (0.6 nmol) or NPY (2.0 nmol) into the third cerebroventricle of male rats during the light cycle. Control animals received vehicle (HPMC/Tween, p.o.) followed 1 h later by ICV vehicle (sterile water). ANOVA indicated significant intergroup differences at all time points. *P < 0.05, *** P < 0.001 versus 0.6 nmol NPY Y5 agonist, not significant (ns) versus 2.0 nmol NPY (n = 5–6). B: Vehicle or NPY5RA-972 (10 mg/kg) was dosed by oral gavage (p.o.) 1 h before administration of NPY Y5 agonist (0.6 nmol) or NPY (0.2 nmol) into the third cerebroventricle of male rats during the light cycle. ANOVA indicated significant intergroup differences at all time points. *P < 0.05, *** P < 0.001 versus 0.6 nmol NPY Y5 agonist, not significant (ns) versus 0.2 nmol NPY (n = 6–7).

Close modal
FIG. 4.

Effect of NPY Y5 selective antagonist NPY5RA-972 on food intake in male Wistar rats induced to feed by previous fasting for 24 h (n = 8–12; A), free-feeding male Wistar rats (n = 8 per group; B), and free-feeding male obese Zucker rats (n = 7–12; C). In each experiment, NPY5RA-972 (1–10 mg/kg) was dosed by oral gavage (p.o.) 1 h before commencement of food intake measurements. In fasted rats, this meant that rats were dosed 1 h before presentation of food. ANOVA indicated no significant intergroup differences at any time point in any of the three experiments.

FIG. 4.

Effect of NPY Y5 selective antagonist NPY5RA-972 on food intake in male Wistar rats induced to feed by previous fasting for 24 h (n = 8–12; A), free-feeding male Wistar rats (n = 8 per group; B), and free-feeding male obese Zucker rats (n = 7–12; C). In each experiment, NPY5RA-972 (1–10 mg/kg) was dosed by oral gavage (p.o.) 1 h before commencement of food intake measurements. In fasted rats, this meant that rats were dosed 1 h before presentation of food. ANOVA indicated no significant intergroup differences at any time point in any of the three experiments.

Close modal
FIG. 5.

Effect of NPY Y5 selective antagonist NPY5RA-972 on food intake induced by the NPY Y5 selective agonist [cPP1–7,NPY19–23, Ala31,Aib32,Gln34]-hPP. Vehicle or NPY5RA-972 (10 mg/kg) was dosed by oral gavage (p.o.) 12 h, or NPY5RA-972 (3 mg/kg p.o.) 1 h before administration of NPY Y5 selective agonist (0.6 nmol) into the third cerebroventricle of male rats. ANOVA indicated significant intergroup differences at both time points. *P < 0.05 versus 0.6 nmol NPY Y5 agonist (n = 5–6).

FIG. 5.

Effect of NPY Y5 selective antagonist NPY5RA-972 on food intake induced by the NPY Y5 selective agonist [cPP1–7,NPY19–23, Ala31,Aib32,Gln34]-hPP. Vehicle or NPY5RA-972 (10 mg/kg) was dosed by oral gavage (p.o.) 12 h, or NPY5RA-972 (3 mg/kg p.o.) 1 h before administration of NPY Y5 selective agonist (0.6 nmol) into the third cerebroventricle of male rats. ANOVA indicated significant intergroup differences at both time points. *P < 0.05 versus 0.6 nmol NPY Y5 agonist (n = 5–6).

Close modal
FIG. 6.

Effect of NPY5RA-972 (10 mg/kg p.o. twice daily) or sibutramine (3 mg/kg, once daily) on food intake (A) and body weight (B) in free-feeding Wistar rats (n = 6–7).

FIG. 6.

Effect of NPY5RA-972 (10 mg/kg p.o. twice daily) or sibutramine (3 mg/kg, once daily) on food intake (A) and body weight (B) in free-feeding Wistar rats (n = 6–7).

Close modal
FIG. 7.

Effect of NPY5RA-972 (10 mg/kg p.o., twice daily) or sibutramine (3 mg/kg, once daily) on food intake (A) and body weight (B) in Wistar rats fed a palatable, high-fat diet. Three groups of rats were fed the palatable diet for 10 weeks before (and throughout) the experiment, and a group of rats fed a standard diet (dosed with only vehicle) were included for comparison (n = 6–8).

FIG. 7.

Effect of NPY5RA-972 (10 mg/kg p.o., twice daily) or sibutramine (3 mg/kg, once daily) on food intake (A) and body weight (B) in Wistar rats fed a palatable, high-fat diet. Three groups of rats were fed the palatable diet for 10 weeks before (and throughout) the experiment, and a group of rats fed a standard diet (dosed with only vehicle) were included for comparison (n = 6–8).

Close modal
TABLE 1

Properties of NPY5RA-972: in vitro profile of NPY Y5 ligands

Binding (IC50, nmol/l)
cAMP reporter response (EC50, nmol/l)cAMP reporter response (IC50, nmol/l)
hNPY Y5hNPY Y1hNPY Y2hNPY Y4rNPY Y5rNPY Y5rNPY Y5 10 nmol/l NPY
NPY 0.11 0.09 0.03 0.47 0.47 0.31 — 
[cPP1–7,NPY19–23,Ala31, Aib32, Gln34]-hPP 0.07 473 14 36 0.22 0.263 — 
NPY5RA-972 3.1 >10,000 >10,000 >10,000 9.3 — 39.2 
Binding (IC50, nmol/l)
cAMP reporter response (EC50, nmol/l)cAMP reporter response (IC50, nmol/l)
hNPY Y5hNPY Y1hNPY Y2hNPY Y4rNPY Y5rNPY Y5rNPY Y5 10 nmol/l NPY
NPY 0.11 0.09 0.03 0.47 0.47 0.31 — 
[cPP1–7,NPY19–23,Ala31, Aib32, Gln34]-hPP 0.07 473 14 36 0.22 0.263 — 
NPY5RA-972 3.1 >10,000 >10,000 >10,000 9.3 — 39.2 

IC50 values at the human NPY Y1, NY Y2, NPY Y4 and NPY Y5 and rat NPY Y5 were determined using membrane binding assays described in research design and methods. Functional activities were determined in cAMP reporter assays using HEK293 cells stably expressing the rat NPY Y5 receptor and a reporter cassette consisting of a cAMP-driven β-galactosidase gene.

TABLE 2

Plasma and CSF concentrations of NPY5RA-972

Oral dose
10 mg/kg (1 h)30 mg/kg (1 h)80 mg/kg (1 h)
n 
Total plasma (μmol/l) 21.7 ± 3.8 35.5 ± 2.8 53.2 ± 7.5 
CSF (μmol/l) 0.153 ± 0.035 0.265 ± 0.033 0.434 ± 0.070 
Oral dose
10 mg/kg (1 h)30 mg/kg (1 h)80 mg/kg (1 h)
n 
Total plasma (μmol/l) 21.7 ± 3.8 35.5 ± 2.8 53.2 ± 7.5 
CSF (μmol/l) 0.153 ± 0.035 0.265 ± 0.033 0.434 ± 0.070 

Compound concentrations in plasma and CSF were determined by high-performance liquid chromatography-ultraviolet (285 nm) analysis.

The technical assistance of Gunn-Britt Forsberg, Britt-Marie Fihn, and Bill Brown and critical evaluation of the manuscript by Drs. David Morgan, David Smith, Lynn Pritchard, and Carl Montague are gratefully acknowledged.

1
Friedman JM: Obesity in the new millennium.
Nature
404
:
632
–634,
2000
2
Horvath TL, Diano S, Sotonyi P, Heiman M, Tschop M: Minireview: ghrelin and the regulation of energy balance—a hypothalamic perspective.
Endocrinology
142
:
4163
–4169,
2001
3
Tritos NA, Maratos-Flier E: Two important systems in energy homeostasis: melanocortins and melanin-concentrating hormone.
Neuropeptides
33
:
339
–349,
1999
4
Kalra SP, Dube MG, Pu S, Xu B, Horvath TL, Kalra PS: Interacting appetite-regulating pathways in the hypothalamic regulation of body weight.
Endocr Rev
20
:
68
–100,
1999
5
Meister B: Control of food intake via leptin receptors in the hypothalamus.
Vitam Horm
59
:
265
–304,
2000
6
Barsh GS, Farooqi IS, O’Rahilly S: Genetics of body-weight regulation.
Nature
404
:
644
–651,
2000
7
Boutin P, Froguel P: Genetics of human obesity.
Best Pract Res Clin Endocrinol Metab
15
:
391
–404,
2001
8
Huszar D, Lynch CA, Fairchild-Huntress V, Dunmore JH, Fang Q, Berkemeier LR, Gu W, Kesterson RA, Boston BA, Cone RD, Smith FJ, Campfield LA, Burn P, Lee F: Targeted disruption of the melanocortin-4 receptor results in obesity in mice.
Cell
88
:
131
–141,
1997
9
Shimada M, Tritos NA, Lowell BB, Flier JS, Maratos-Flier E: Mice lacking melanin-concentrating hormone are hypophagic and lean.
Nature
396
:
670
–674,
1998
10
Yaswen L, Diehl N, Brennan MB, Hochgeschwender U: Obesity in the mouse model of pro-opiomelanocortin deficiency responds to peripheral melanocortin.
Nat Med
5
:
1066
–1070,
1999
11
Beck B: KO’s and organisation of peptidergic feeding behavior mechanisms.
Neurosci Biobehav Rev
25
:
143
–158,
2001
12
Wieland HA, Hamilton BS, Krist B, Doods HN: The role of NPY in metabolic homeostasis: implications for obesity therapy.
Expert Opin Investig Drugs
9
:
1327
–1346,
2000
13
Zimanyi IA, Poindexter GS: NPY-ergic agents for the treatment of obesity.
Drug Dev Res
51
:
94
–111,
2000
14
Erickson JC, Clegg KE, Palmiter RD: Sensitivity to leptin and susceptibility to seizures of mice lacking neuropeptide Y.
Nature
381
:
415
–421,
1996
15
Erickson JC, Hollopeter G, Palmiter RD: Attenuation of the obesity syndrome of ob/ob mice by the loss of neuropeptide Y.
Science
274
:
1704
–1707,
1996
16
Inui A: Neuropeptide Y feeding receptors: are multiple subtypes involved?
Trends Pharmacol Sci
20
:
43
–46,
1999
17
Gerald C, Walker MW, Criscione L, Gustafson EL, Batzl-Hartmann C, Smith KE, Vaysse P, Durkin MM, Laz TM, Linemeyer DL, Schaffhauser AO, Whitebread S, Hofbauer KG, Taber RI, Branchek TA, Weinshank RL: A receptor subtype involved in neuropeptide-Y-induced food intake.
Nature
382
:
168
–171,
1996
18
Hu Y, Bloomquist BT, Cornfield LJ, DeCarr LB, Flores-Riveros JR, Friedman L, Jiang P, Lewis-Higgins L, Sadlowski Y, Schaefer J, Velazquez N, McCaleb ML: Identification of a novel hypothalamic neuropeptide Y receptor associated with feeding behavior.
J Biol Chem
271
:
26315
–26319,
1996
19
Haynes AC, Arch JR, Wilson S, McClue S, Buckingham RE: Characterisation of the neuropeptide Y receptor that mediates feeding in the rat: a role for the Y5 receptor?
Regul Pept
75–76
:
355
–361,
1998
20
Cabrele C, Langer M, Bader R, Wieland HA, Doods HN, Zerbe O, Beck-Sickinger AG: The first selective agonist for the neuropeptide YY5 receptor increases food intake in rats.
J Biol Chem
275
:
36043
–36048,
2000
21
McCrea K, Wisialowski T, Cabrele C, Church B, Beck-Sickinger A, Kraegen E, Herzog H: 2–36[K4,RYYSA(19–23)]PP a novel Y5-receptor preferring ligand with strong stimulatory effect on food intake.
Regul Pept
87
:
47
–58,
2000
22
Parker EM, Balasubramaniam A, Guzzi M, Mullins DE, Salisbury BG, Sheriff S, Witten MB, Hwa JJ: [D-Trp(34)] neuropeptide Y is a potent and selective neuropeptide Y Y(5) receptor agonist with dramatic effects on food intake.
Peptides
21
:
393
–399,
2000
23
Wyss P, Stricker-Krongrad A, Brunner L, Miller J, Crossthwaite A, Whitebread S, Criscione L: The pharmacology of neuropeptide Y (NPY) receptor-mediated feeding in rats characterizes better Y5 than Y1, but not Y2 or Y4 subtypes.
Regul Pept
75–76
:
363
–371,
1998
24
Polidori C, Ciccocioppo R, Regoli D, Massi M: Neuropeptide Y receptor(s) mediating feeding in the rat: characterization with antagonists.
Peptides
21
:
29
–35,
2000
25
Campbell RE, ffrench-Mullen JM, Cowley MA, Smith MS, Grove KL: Hypothalamic circuitry of neuropeptide Y regulation of neuroendocrine function and food intake via the Y5 receptor subtype.
Neuroendocrinology
74
:
106
–119,
2001
26
Flynn MC, Turrin NP, Plata-Salaman CR, ffrench-Mullen JM: Feeding response to neuropeptide Y-related compounds in rats treated with Y5 receptor antisense or sense phosphothio-oligodeoxynucleotide.
Physiol Behav
66
:
881
–884,
1999
27
Tang-Christensen M, Kristensen P, Stidsen CE, Brand CL, Larsen PJ: Central administration of Y5 receptor antisense decreases spontaneous food intake and attenuates feeding in response to exogenous neuropeptide Y.
J Endocrinol
159
:
307
–312,
1998
28
Schaffhauser AO, Stricker-Krongrad A, Brunner L, Cumin F, Gerald C, Whitebread S, Criscione L, Hofbauer KG: Inhibition of food intake by neuropeptide Y Y5 receptor antisense oligodeoxynucleotides.
Diabetes
46
:
1792
–1798,
1997
29
Marsh DJ, Hollopeter G, Kafer KE, Palmiter RD: Role of the Y5 neuropeptide Y receptor in feeding and obesity.
Nat Med
4
:
718
–721,
1998
30
Criscione L, Rigollier P, Batzl-Hartmann C, Rueger H, Stricker-Krongrad A, Wyss P, Brunner L, Whitebread S, Yamaguchi Y, Gerald C, Heurich RO, Walker MW, Chiesi M, Schilling W, Hofbauer KG, Levens N: Food intake in free-feeding and energy-deprived lean rats is mediated by the neuropeptide Y5 receptor.
J Clin Invest
102
:
2136
–2145,
1998
31
Block MH, Boyer S, Brailsford W, Brittain D, Carrol D, Capman S, Clarke D, Donald C, Foote K, Godfrey L, Ladner T, Marsham P, Masters D, Mee CD, O’Donovan M, Pease JE, Pickup A, Rayner J, Roberts A, Schofield P, Suleman A, Turnbull AV: The discovery and optimization of a series of carbazole ureas as NPY Y5 antagonists for the treatment of obesity.
J Med Chem
In press,
2002
32
Paxinos G, Watson C:
The Rat Brain in Stereotaxic Co-ordinates.
San Diego, CA, Academic Press,
1998
33
Stock MJ: Sibutramine: a review of the pharmacology of a novel anti-obesity agent.
Int J Obes Relat Metab Disord
21 (Suppl. 1)
:
S25
–S29,
1997
34
Brown M, Bing C, King P, Pickavance L, Heal D, Wilding J: Sibutramine reduces feeding, body fat and improves insulin resistance in dietary-obese male Wistar rats independently of hypothalamic neuropeptide Y.
Br J Pharmacol
132
:
1898
–1904,
2001
35
Levin BE, Dunn-Meynell AA: Defense of body weight against chronic caloric restriction in obesity-prone and -resistant rats.
Am J Physiol Regul Integr Comp Physiol
278
:
R231
–R237,
2000
36
Levin BE, Dunn-Meynell AA, Balkan B, Keesey RE: Selective breeding for diet-induced obesity and resistance in Sprague-Dawley rats.
Am J Physiol
273
:
R725
–R730,
1997
37
Boozer CN, Leibel RL, Love RJ, Cha MC, Aronne LJ: Synergy of sibutramine and low-dose leptin in treatment of diet-induced obesity in rats.
Metabolism
50
:
889
–893,
2001
38
Zimanyi IA, Ortiz AA, Rassnick S, Hogan JB, Huang Y, Bruce MA, Gillman KW, Poindexter GS:
Evidence Supporting the Role of the Y1, but not the Y5 Receptor in Food Intake in Rats
(Abstract). Presented at the 6th International NPY Conference, Sydney, Australia,
22
–26 April
2001
39
Nichol KA, Morey A, Couzens MH, Shine J, Herzog H, Cunningham AM: Conservation of expression of neuropeptide Y5 receptor between human and rat hypothalamus and limbic regions suggests an integral role in central neuroendocrine control.
J Neurosci
19
:
10295
–10304,
1999
40
Jacques D, Tong Y, Shen SH, Quirion R: Discrete distribution of the neuropeptide Y Y5 receptor gene in the human brain: an in situ hybridization study.
Brain Res Mol Brain Res
61
:
100
–107,
1998
41
Jenkinson CP, Cray K, Walder K, Herzog H, Hanson, Ravussin E: Novel polymorphisms in the neuropeptide-Y Y5 receptor associated with obesity in Pima Indians.
Int J Obes Relat Metab Disord
24
:
580
–584,
2000
42
Roche C, Boutin P, Dina C, Gyapay G, Basdevant A, Hager J, Guy-Grand B, Clement K, Froguel P: Genetic studies of neuropeptide Y and neuropeptide Y receptors Y1 and Y5 regions in morbid obesity.
Diabetologia
40
:
671
–675,
1997
43
Rosenkranz K, Hinney A, Ziegler A, von Prittwitz S, Barth N, Roth H, Mayer H, Siegfried W, Lehmkuhl G, Poustka F, Schmidt M, Schafer H, Remschmidt H, Hebebrand J: Screening for mutations in the neuropeptide Y Y5 receptor gene in cohorts belonging to different weight extremes.
Int J Obes Relat Metab Disord
22
:
157
–163,
1998
44
Zuana OD, Sadlo M, Germain M, Feletou M, Chamorro S, Tisserand F, Montrion C, Boivin JF, Duhault J, Boutin JA, Levens N: Reduced food intake in response to CGP 71683A may be due to mechanisms other than NPY Y5 receptor blockade.
Int J Obes Relat Metab Disord
25
:
84
–94,
2001
45
Bannon AW, Seda J, Carmouche M, Francis JM, Norman MH, Karbon B, McCaleb ML: Behavioral characterization of neuropeptide Y knockout mice.
Brain Res
868
:
79
–87,
2000
46
Criscione L, Schaffhauser AO, Guerini D, Rigollier P, Schmid T, Buhlmayer P:
Effects of the Y5 Receptor Antagonist CGP71683A on Food Intake in Male Y5 Receptor Knock-Out Mice
(Abstract). Presented at the 6th International NPY Conference, Sydney, Australia,
22
–26 April
2001
47
Kordik CP, Luo C, Zanoni BC, Lovenberg TW, Wilson SJ, Vaidya AH, Crooke JJ, Rosenthal DI, Reitz AB: Pyrazolecarboxamide human neuropeptide Y5 receptor ligands with in vivo antifeedant activity.
Bioorg Med Chem Lett
11
:
2287
–2290,
2001
48
Duhault J, Boulanger M, Chamorro S, Boutin JA, Della ZO, Douillet E, Fauchere JL, Feletou M, Germain M, Husson B, Vega AM, Renard P, Tisserand F: Food intake regulation in rodents: Y5 or Y1 NPY receptors or both?
Can J Physiol Pharmacol
78
:
173
–185,
2000
49
Daniels AJ, Grizzle MK, Matthews JE, Wiard R, Heyer D, Burnette T, Koegler F, Cameron JL:
The Role of NPY-Y5 Receptors in Feeding Probed by Novel Y5 Antagonists
(Abstract). Presented at the Society for Neuroscience’s 31st Annual Meeting, San Diego, CA,
10
–15 November,
2001
50
Elliott RL, Oliver RM, Hammond M, Patterson PA, She L, Hargrove DM, Martin KA, Maurer TS, Kalvass JC, Morgan BM, DaSilva-Jardine P, Stevenson RW, Mack C, Cassella J:
The In Vitro and In Vivo Characterisation of 3-{2-[6-(2-tert-butoxy-ethoxy)-pyridin-3-yl]-1H-imidazol-4-yl}-benzonitrile, (I), a Potent and Selective NPY-Y5 Antagonist
(Abstract). Presented at the 6th International NPY Conference, Sydney, Australia,
22
–26 April 2001
51
Kanatani A, Ishihara A, Iwaasa H, Nakamura K, Okamoto O, Hidaka M, Ito J, Fukuroda T, MacNeil DJ, Van der Ploeg LH, Ishii Y, Okabe T, Fukami T, Ihara M: L-152,804: orally active and selective neuropeptide Y Y5 receptor antagonist.
Biochem Biophys Res Commun
272
:
169
–173,
2000
52
Kanatani A, Fukami T, Ishihara A, Ishii Y, MacNeil DJ, Van der Ploeg LH, Ihara M:
Anorexigenic Effects of Orally-Active NPY Antagonists: Participation of Y1 and Y5 Receptors in Feeding Behaviour
(Abstract). Presented at the 6th International NPY Conference, Sydney, Australia,
2001
53
O’Shea D, Morgan DG, Meeran K, Edwards CM, Turton MD, Choi SJ, Heath MM, Gunn I, Taylor GM, Howard JK, Bloom CI, Small CJ, Haddo O, Ma JJ, Callinan W, Smith DM, Ghatei MA, Bloom SR: Neuropeptide Y induced feeding in the rat is mediated by a novel receptor.
Endocrinology
138
:
196
–202,
1997
54
Iyengar S, Li DL, Simmons RM: Characterization of neuropeptide Y-induced feeding in mice: do Y1–Y6 receptor subtypes mediate feeding?
J Pharmacol Exp Ther
289
:
1031
–1040,
1999
55
Kanatani A, Mashiko S, Murai N, Sugimoto N, Ito J, Fukuroda T, Fukami T, Morin N, MacNeil DJ, Van der Ploeg LH, Saga Y, Nishimura S, Ihara M: Role of the Y1 receptor in the regulation of neuropeptide Y-mediated feeding: comparison of wild-type, Y1 receptor-deficient, and Y5 receptor-deficient mice.
Endocrinology
141
:
1011
–1016,
2000
56
Marsh DJ, Baraban SC, Hollopeter G, Palmiter RD: Role of the Y5 neuropeptide Y receptor in limbic seizures.
Proc Natl Acad Sci U S A
96
:
13518
–13523,
1999

Address correspondence and reprint requests to Dr. Andrew V. Turnbull, Cardiovascular and Gastrointestinal Discovery Department, AstraZeneca, Mereside, Alderley Park, Macclesfield, SK4 10TG, U.K. E-mail: andrew.turnbull@astrazeneca.com.

Received for publication 20 December 2001 and accepted in revised form 29 April 2002.

CNS, central nervous system; CSF, cerebrospinal fluid; DMEM, Dulbecco’s modified Eagle’s medium; HPMC, hydroxypropylmethylcellulose; ICV, intracerebroventricular; NPY, neuropeptide Y.