OBJECTIVE

Glucagon-like peptide (GLP)-1 is a regulatory peptide synthesized in the gut and the brain that plays an important role in the regulation of food intake. Both GLP-1 and exendin (Ex)-4, a long-acting GLP-1 receptor (GLP-1r) agonist, reduce food intake when administered intracerebroventricularly, whereas Ex4 is much more potent at suppressing food intake when given peripherally. It has generally been hypothesized that this difference is due to the relative pharmacokinetic profiles of GLP-1 and Ex4, but it is possible that the two peptides control feeding via distinct mechanisms.

RESEARCH DESIGN AND METHODS

In this study, the anorectic effects of intracerebroventricular GLP-1 and Ex4, and the sensitivity of these effects to GLP-1r antagonism, were compared in rats. In addition, the GLP-1r dependence of the anorectic effect of intracerebroventricular Ex4 was assessed in GLP-1r−/− mice.

RESULTS

Intracerebroventricular Ex4 was 100-fold more potent than GLP-1 at reducing food intake, and this effect was insensitive to GLP-1r antagonism. However, GLP-1r antagonists completely blocked the anorectic effect of intraperitoneal Ex4. Despite the insensitivity of intracerebroventricular Ex4 to GLP-1r antagonism, intracerebroventricular Ex4 failed to reduce food intake in GLP-1r−/− mice.

CONCLUSIONS

These data suggest that although GLP-1rs are required for the actions of Ex4, there appear to be key differences in how GLP-1 and Ex4 interact with central nervous system GLP-1r and in how Ex4 interacts with GLP-1r in the brain versus the periphery. A better understanding of these unique differences may lead to expansion and/or improvement of GLP-1–based therapies for type 2 diabetes and obesity.

Glucagon-like peptide (GLP)-1 is a product of the preproglucagon gene (1) that is synthesized in the distal ileum (2) as well as the caudal nucleus of the solitary tract (NTS) and ventrolateral medulla (3). Although GLP-1 is perhaps best known for its essential role in the regulation of peripheral glucose homeostasis, multiple lines of evidence suggest that GLP-1 also acts in the central nervous system (CNS) to regulate food intake. In support of this hypothesis, long-acting GLP-1 receptors (GLP-1rs) are expressed in brain regions known to regulate energy balance, such as the mediobasal hypothalamus and the caudal brainstem (3,4), and consistent with a role for GLP-1 as a putative satiety signal, central administration of GLP-1 potently reduces short-term food intake (5,6). Conversely, central administration of the GLP-1r antagonist exendin (Ex) (9-39) (Ex9) increases food intake and body weight (7), suggesting that endogenous GLP-1 has a physiological role in the regulation of energy balance.

Recently, the GLP-1 system has emerged as a novel therapeutic target for type 2 diabetes, as peripheral GLP-1 infusion effectively lowers blood glucose levels and improves glucose tolerance in humans (8). However, because circulating active GLP-1 is rapidly degraded by the enzyme dipeptidyl peptidase-4 (DPP-4) (9,11), alternative strategies for targeting the GLP-1 system have been developed, including stable GLP-1 analogues and DPP-4 inhibitors. One such analog is Ex4, a peptide originally isolated from the saliva of the Gila monster (Heloderma suspectum), which is a highly potent, DPP-4–resistant GLP-1r agonist in vitro and in vivo (12,13). Recently, exenatide (a synthetic Ex4) and the DPP-4 inhibitor sitagliptin were Food and Drug Administration approved as therapies for type 2 diabetes. However, whereas both drugs effectively improved glycemic control in clinical trials (14,15), Ex4, but not sitagliptin, was also associated with significant weight loss (14,16).

The above finding is compelling in that it raises the possibility that Ex4, at doses used clinically, may have in vivo actions that are substantively different from those of intact GLP-1 achieved through DPP-4 inhibition. Although studies using GLP-1r knockout (GLP-1r−/−) mice provide strong evidence that the GLP-1r is necessary for the in vivo actions of Ex4 (17,,,21), other studies using GLP-1r antagonists suggest that Ex4, particularly in the brain, may act, at least in part, independently of GLP-1r (22,24). Therefore, we tested the hypothesis that the central anorectic effect of Ex4 is different from that of GLP-1.

Adult male Long-Evans rats (Harlan, Indianapolis, IN), GLP-1r−/− mice, and their wild-type C57BL/6J littermates were housed individually in plastic rodent cages and maintained on a 12-h light/dark cycle with ad libitum access to water and pelleted rodent diet (Harlan Teklad). Rats and mice were outfitted with cannulas (Plastics One, Roanoake, VA) aimed at the third cerebral ventricle, and correct cannula placement was verified as previously described (25,26). All procedures were approved by the University of Cincinnati Institutional Animal Care and Use Committee.

Peptides.

GLP-1 and Ex4 were obtained from Bio Nebraska (Lincoln, NE) and American Peptide (Sunnyvale, CA), respectively. The GLP-1r antagonists His1, Glu8 Ex4 (dHEx), and Ex (9-39) (Ex9) were obtained from Baylor College of Medicine Protein Synthesis Core (Houston, TX) and Tocris (Ellisville, MO), respectively. All peptides were dissolved in saline and administered either intracerebroventricularly in a volume of 1.0 μl or intraperitoneally in a volume of 1.0 ml/kg.

Food intake studies.

Rats and mice were fed ad libitum at all times except for the mornings of study days. During this time, food was removed from the animals' cages and weighed 4 h before lights off, and animals were assigned to weight-matched groups. Pretreatment (saline or GLP-1r antagonist) injections commenced 1 h before lights off, and treatment (saline or GLP-1r agonist) injections commenced 30 min before lights off. Injection order was counterbalanced across all experimental groups to evenly distribute subtle variations in timing of injections. Food was returned to the animals' cages at lights off, and food intake and body weight were measured at selected time points.

First, dose-response curves for anorexia induced by intracerebroventricular GLP-1 (0, 0.3, 1.0, 3.0, and 10.0 μg) and Ex4 (0, 0.01, 0.03, 0.1, and 0.3 μg) were established. Based on these results, time courses of anorexia induced by intracerebroventricular GLP-1 (3.0 nmol, ∼10.0 μg) and Ex4 (0.03 nmol, ∼0.1 μg) were compared, and conditioned taste aversion (CTA) to the same doses of intracerebroventricular GLP-1 and Ex4 was assessed as previously described (27).

To assess the ability of central GLP-1r antagonism to block anorexia induced by central GLP-1 and Ex4, rats were pretreated with intracerebroventricular saline, dHEx (10.0 μg, [28]), or Ex9 (100.0 μg) and then treated with intracerebroventricular saline, GLP-1 (10.0 μg), or Ex4 (0.1 μg). To assess the ability of peripheral GLP-1r antagonism to block anorexia induced by peripheral Ex4, rats were pretreated with intraperitoneal saline or dHEx (1.0 mg/kg) and then treated with intraperitoneal saline or Ex4 (10.0 μg/kg). Finally, to assess the ability of central Ex4 to reduce food intake in GLP-1r−/− mice, mice were treated with intracerebroventricular saline or Ex4 (1.0 μg).

c-Fos immunohistochemistry.

To assess the ability of central GLP-1r antagonism to block neuronal activation induced by central GLP-1 and Ex4, rats were pretreated with intracerebroventricular saline or dHEx (10.0 μg) and then treated with intracerebroventricular saline, GLP-1 (10.0 μg), or Ex4 (0.1 μg). Two hours later, rats were deeply anesthetized with sodium pentobarbital and perfused transcardially with 0.1 mol/l PBS followed by 4.0% paraformaldehyde/PBS. Brains were postfixed at 4°C for 24 h in 4.0% paraformaldehyde/PBS and stored at 4°C in 30.0% sucrose/PBS. Serial coronal forebrain sections and longitudinal hindbrain sections were collected at 35 μm using a freezing microtome and stored at −20°C in cryoprotectant.

After washing with PBS, sections were incubated in 1.0% hydrogen peroxide/PBS for 10 min, followed by 1.0% sodium borohydride/PBS for 30 min. Sections were blocked for 1 h in 0.1% BSA/0.4% Triton-X-100/PBS and incubated overnight at room temperature in blocking solution containing rabbit anti–c-Fos diluted at 1:5,000 (sc-52; Santa Cruz Biotechnology, Santa Cruz, CA). The next morning, sections were washed and incubated at room temperature for 1 h in blocking solution containing biotinylated goat anti-rabbit IgG diluted at 1:200 (BA-1000; Vector Laboratories, Burlingame, CA) followed by 1 h in ABC solution diluted 1:800 in PBS (PK6100; Vector Laboratories) and 10 min in DAB-nickel solution. Finally, sections were washed with 0.1 mol/l phosphate buffer, mounted on gelatin-coated slides, and cover slipped.

For quantification of c-Fos immunoreactivity in the central nucleus of the amygdala (CeA), paraventricular nucleus of the hypothalamus (PVN), and nucleus of the solitary tract (NTS), digital images of sections were acquired using a digital camera attached to a Zeiss microscope (Zeiss, Thornwood, NY). For each brain, two sections per area were analyzed, and special care was taken to compare only sections within the same plane along the rostro-caudal (CeA and PVN) or dorso-ventral (NTS) axis. c-Fos immunoreactivity was quantified as optical density using the National Institutes of Health program Scion Image.

Tissue culture studies.

INS-1 cells were seeded in 35-mm six-well plates at a density of 2 × 105 cell/well in 1.5 ml of media consisting of RPMI-1640 supplemented with 10% heat-inactivated fetal bovine serum (FBS), 1.0 mmol/l sodium pyruvate, 2.0 mmol/l l-glutamine, 50.0 μmol/l β-mercaptoethanol, and 0.5 mg/ml gentamicin sulfate and grown in a 37°C incubator in an atmosphere of 5% CO2 and 95% air and 100% humidity for 3 days until nearly confluent. On day 4, cells were washed with PBS and replaced with fresh media. On day 5, cells were preincubated for 2 h in 2.0 ml of buffer consisting of Krebs-Ringer bicarbonate buffer (KRB) supplemented with 0.1% BSA and 30 mg/dl glucose and then washed twice with 2.0 ml of the same buffer solution. Cells were then incubated for 1 h in 1.0 ml of KRB supplemented with 0.1% BSA, 200 mg/dl glucose, and 1.0 nmol/l GLP-1, 0.01 nmol/l Ex4, or 1.0 nmol/l Ex4 with or without 100 nmol/l dHEx. Finally, incubation buffer was harvested, centrifuged, decanted, and stored at −20°C for immunoreactive insulin (IRI) assay, and cells were washed once with 1.0 ml of preincubation buffer and then extracted with 1.0 ml of acid ethanol for 2 h at −20°C, after which acid ethanol was diluted 1:200 with Tris assay buffer for IRI assay in cell layer. IRI was measured using a radioimmunoassay as previously described (29).

Statistical analysis.

All values are reported as means ± SE. Data were analyzed using one- or two-way ANOVA or two-way repeated-measures ANOVA. Post hoc multiple comparisons were made using Tukey's post hoc test. Significance was set at P < 0.05 for all analyses.

Comparison of intracerebroventricular GLP-1– and Ex4-induced anorexia.

Consistent with previous reports, intracerebroventricular GLP-1 and Ex4 elicited potent, dose-dependent reductions in 4-h food intake (Fig. 1 A and B; P < 0.05, one-way ANOVA with Tukey's post hoc test). However, Ex4 significantly reduced food intake at doses much lower than those of GLP-1. Specifically, 10.0 μg of GLP-1 and 0.1 μg of Ex4 produced comparable degrees of anorexia, reducing food intake to 56 and 45% of control values, respectively. These data indicate that, when administered into the third ventricle, Ex4 is roughly 100-fold more potent than GLP-1 at reducing food intake.

FIG. 1.

Comparison of anorectic effects of intracerebroventricular GLP-1 and Ex4. A and B: Dose-response curves for intracerebroventricular GLP-1 (A) and Ex4 (B). Cumulative 4-h food intake is shown. C: Time course of anorectic effects of intracerebroventricular GLP-1 (3.0 nmol) and Ex4 (0.03 nmol) over 24 h. □, Saline; ▨, GLP-1 (3.0 nmol/l); , Ex4 (0.03 nmol/l). D: Preference ratios for 0.1% saccharin versus total fluid intake during the 4-h two-bottle access to saccharin and water. Saccharin was previously paired with intracerebroventricular saline, intracerebroventricular GLP-1 (3.0 nmol), intracerebroventricular Ex4 (0.03 nmol), or intraperitoneal LiCl (0.15 mol/l administered at 2.0% body wt). Dotted line represents preference ratio for saline-treated rats. Data are represented as means ± SE. *P < 0.05 vs. saline. #P < 0.05 vs. GLP-1.

FIG. 1.

Comparison of anorectic effects of intracerebroventricular GLP-1 and Ex4. A and B: Dose-response curves for intracerebroventricular GLP-1 (A) and Ex4 (B). Cumulative 4-h food intake is shown. C: Time course of anorectic effects of intracerebroventricular GLP-1 (3.0 nmol) and Ex4 (0.03 nmol) over 24 h. □, Saline; ▨, GLP-1 (3.0 nmol/l); , Ex4 (0.03 nmol/l). D: Preference ratios for 0.1% saccharin versus total fluid intake during the 4-h two-bottle access to saccharin and water. Saccharin was previously paired with intracerebroventricular saline, intracerebroventricular GLP-1 (3.0 nmol), intracerebroventricular Ex4 (0.03 nmol), or intraperitoneal LiCl (0.15 mol/l administered at 2.0% body wt). Dotted line represents preference ratio for saline-treated rats. Data are represented as means ± SE. *P < 0.05 vs. saline. #P < 0.05 vs. GLP-1.

Close modal

Figure 1,C illustrates the time course of intracerebroventricular GLP-1– and Ex4-induced anorexia. Whereas 3.0 nmol (∼10.0 μg) of GLP-1 and 0.03 nmol (∼0.1 μg) of Ex4 both actively suppressed food intake up to 4 h, only Ex4 elicited persistent anorexia that remained detectable throughout the 24 h of observation (P < 0.05, two-way repeated-measures ANOVA with Tukey's post hoc test). Furthermore, these doses of GLP-1 and Ex4 both led to the formation of a CTA (Fig. 1 D; P < 0.05, one-way ANOVA with Tukey's post hoc test). Interestingly, there was a strong trend toward a significantly lower preference ratio of Ex4-treated rats versus GLP-1–treated rats (P = 0.052), suggesting that the aversive effects of Ex4 were more pronounced than those of GLP-1.

Sensitivity of intracerebroventricular GLP-1 and Ex4 to GLP-1r antagonism.

Although previous studies have reported an inability to block certain effects of Ex4 with GLP-1r antagonists, these studies did not necessarily account for the significantly greater potency of Ex4 over GLP-1. Therefore, we sought to compare the ability of GLP-1r antagonists to block anorexia and neuronal activation induced by doses of intracerebroventricular GLP-1 and Ex4 that produce effects of comparable magnitude. Pretreatment with either 10.0 μg of dHEx or 100.0 μg of Ex9 caused near-complete blockade of anorexia induced by 10.0 μg of GLP-1 (Fig. 2,A and C; P < 0.05 by two-way ANOVA with Tukey's post hoc test). However, whereas 0.1 μg of Ex4 and 10.0 μg of GLP-1 elicited comparable degrees of anorexia, the doses of dHEx and Ex9 that nearly abolished GLP-1–induced anorexia failed to block the anorectic effect of Ex4 (Fig. 2 B and D), although a nonsignificant trend was observed with dHEx (P = 0.148).

FIG. 2.

Effect of GLP-1r antagonists on anorexia induced by intracerebroventricular GLP-1 and Ex4. A and B: Rats were pretreated with intracerebroventricular dHEx (10 μg) followed by intracerebroventricular GLP-1 (10 μg) (A) or Ex4 (0.1 μg) (B). C and D: Rats were pretreated with intracerebroventricular Ex9 (100 μg) followed by intracerebroventricular GLP-1 (10 μg) (C) or Ex4 (0.1 μg) (D). Cumulative 4-h food intake is shown. Data are represented as means ± SE. *P < 0.05 vs. Sal/Sal. #P < 0.05 vs. Sal/GLP-1.

FIG. 2.

Effect of GLP-1r antagonists on anorexia induced by intracerebroventricular GLP-1 and Ex4. A and B: Rats were pretreated with intracerebroventricular dHEx (10 μg) followed by intracerebroventricular GLP-1 (10 μg) (A) or Ex4 (0.1 μg) (B). C and D: Rats were pretreated with intracerebroventricular Ex9 (100 μg) followed by intracerebroventricular GLP-1 (10 μg) (C) or Ex4 (0.1 μg) (D). Cumulative 4-h food intake is shown. Data are represented as means ± SE. *P < 0.05 vs. Sal/Sal. #P < 0.05 vs. Sal/GLP-1.

Close modal

To determine whether neuronal activation in response to GLP-1 and Ex4 was also differentially sensitive to GLP-1r antagonism, the effect of intracerebroventricular dHEx to block c-Fos immunoreactivity induced by intracerebroventricular GLP-1 and Ex4 was compared. At the same doses as used above, GLP-1 and Ex4 both induced c-Fos immunoreactivity in identical brain regions, including the CeA, PVN, and the NTS (Fig. 3 A–C; P < 0.05 by two-way ANOVA with Tukey's post hoc test). The magnitude of c-Fos immunoreactivity induced by GLP-1 and Ex4 was similar in the PVN and NTS, whereas GLP-1 induced slightly more c-Fos immunoreactivity than Ex4 in the CeA. In the CeA, dHEx significantly blocked c-Fos immunoreactivity induced by GLP-1 (P < 0.05); however, in the PVN and the NTS, this difference failed to reach statistical significance. Nonetheless, for all three regions, the amount of c-Fos immunoreactivity in brains treated with dHEx and Ex4 was significantly greater than that of brains treated with saline, dHEx alone, or dHEx and GLP-1 (P < 0.05). These results, combined with the food intake data, suggest that CNS actions of Ex4 are relatively insensitive to competitive GLP-1r antagonism.

FIG. 3.

Effect of dHEx on c-Fos immunoreactivity induced by intracerebroventricular GLP-1 and Ex4. Quantification of c-Fos–positive nuclei in the PVN (A), NTS (B), and CeA (C) of rats that were treated with intracerebroventricular saline or dHEx (10 μg) followed by intracerebroventricular saline, GLP-1 (10 μg), or Ex4 (0.1 μg) and killed 2 h later. Data are represented as means ± SE. *P < 0.05 vs. Sal/Sal. #P < 0.05 vs. Sal/GLP-1.

FIG. 3.

Effect of dHEx on c-Fos immunoreactivity induced by intracerebroventricular GLP-1 and Ex4. Quantification of c-Fos–positive nuclei in the PVN (A), NTS (B), and CeA (C) of rats that were treated with intracerebroventricular saline or dHEx (10 μg) followed by intracerebroventricular saline, GLP-1 (10 μg), or Ex4 (0.1 μg) and killed 2 h later. Data are represented as means ± SE. *P < 0.05 vs. Sal/Sal. #P < 0.05 vs. Sal/GLP-1.

Close modal

Potency of Ex4 and sensitivity to GLP-1r antagonism in vitro.

Because dHEx, a validated but lesser used GLP-1r antagonist (28,30,31), failed to block anorexia and neuronal activation induced by intracerebroventricular Ex4, we sought to determine whether dHEx is an effective antagonist of Ex4 in vitro by assessing its ability to block insulin secretion induced by Ex4 in the rat pancreatic islet cell line INS-1. As expected, 1.0 nmol/l GLP-1 significantly augmented insulin secretion above that of glucose alone, and this effect was completely blocked by coincubation with 100 nmol/l dHEx (Fig. 4; P < 0.05 by two-way ANOVA with Tukey's post hoc test). However, in contrast to our in vivo data, 0.01 nmol/l Ex4 failed to augment insulin secretion, whereas 1.0 nmol/l Ex4 had an effect that was comparable to 1.0 nmol/l GLP-1. Moreover, this effect was completely blocked by coincubation with 100 nmol/l dHEx (P < 0.05).

FIG. 4.

Effect of dHEx (100 nmol/l) on insulin secretion induced by GLP-1 (1.0 nmol/l) and Ex4 (0.01 and 1.0 nmol/l) in the presence of glucose (200 mg%). Data are represented as means ± SE. *P < 0.05 vs. glucose. #P < 0.05 vs. glucose + GLP-1 (1.0 nmol/l) or glucose + Ex4 (1.0 nmol/l). □, Saline; ■, 100 nmol/l dHEx.

FIG. 4.

Effect of dHEx (100 nmol/l) on insulin secretion induced by GLP-1 (1.0 nmol/l) and Ex4 (0.01 and 1.0 nmol/l) in the presence of glucose (200 mg%). Data are represented as means ± SE. *P < 0.05 vs. glucose. #P < 0.05 vs. glucose + GLP-1 (1.0 nmol/l) or glucose + Ex4 (1.0 nmol/l). □, Saline; ■, 100 nmol/l dHEx.

Close modal

Sensitivity of intraperitoneal Ex4 to GLP-1r antagonism.

To determine whether the insensitivity of Ex4 to GLP-1r antagonism was specific to CNS administration, we assessed the ability of dHEx to block anorexia induced by intraperitoneal Ex4. As expected, 10 μg/kg of intraperitoneal Ex4 significantly reduced food intake at 4 h (Fig. 5; P < 0.05 by two-way ANOVA with Tukey's post hoc test). Surprisingly, pretreatment with 1.0 mg/kg i.p. dHEx, the same 100-fold excess of antagonist that failed to block anorexia induced by intracerebroventricular Ex4, significantly attenuated this effect (P < 0.05).

FIG. 5.

Effect of intraperitoneal dHEx (1.0 mg/kg) on anorexia induced by intraperitoneal Ex4 (10.0 μg/kg). Cumulative 4-h food intake is shown. Data are represented as means ± SE. *P < 0.05 vs. Sal/Sal. #P < 0.05 vs. Sal/Ex4.

FIG. 5.

Effect of intraperitoneal dHEx (1.0 mg/kg) on anorexia induced by intraperitoneal Ex4 (10.0 μg/kg). Cumulative 4-h food intake is shown. Data are represented as means ± SE. *P < 0.05 vs. Sal/Sal. #P < 0.05 vs. Sal/Ex4.

Close modal

Effect of intracerebroventricular Ex4 in wild-type and GLP-1r−/− mice.

The insensitivity of CNS Ex4 effects to GLP-1r antagonism raises the possibility that Ex4 may act in part via a GLP-1r–independent mechanism. To determine whether the GLP-1r is required for the central anorectic effect of Ex4, intracerebroventricular Ex4 was administered to wild-type and GLP-1r−/− mice. In wild-type mice, 1.0 μg of intracerebroventricular Ex4 elicited profound anorexia such that daily food intake and body weight were significantly reduced for up to 48 and 72 h, respectively (Fig. 6,A and B; P < 0.05 by two-way repeated-measures ANOVA with Tukey's post hoc test). Conversely, this same high dose of intracerebroventricular Ex4 had no effect on food intake or body weight in GLP-1r−/− mice (Fig. 6 C and D).

FIG. 6.

Effect of intracerebroventricular Ex4 in wild-type and GLP-1r−/− mice. A and B: Wild-type mice received intracerebroventricular saline (□) or Ex4 (1.0 μg) (■). Food intake (A) and body weight change (B) were measured over 96 h. C and D: GLP-1r−/− mice received intracerebroventricular saline or Ex4 (1.0 μg). Food intake (C) and body weight change (D) were measured over 24 h. Data are represented as means ± SE. *P < 0.05 vs. saline.

FIG. 6.

Effect of intracerebroventricular Ex4 in wild-type and GLP-1r−/− mice. A and B: Wild-type mice received intracerebroventricular saline (□) or Ex4 (1.0 μg) (■). Food intake (A) and body weight change (B) were measured over 96 h. C and D: GLP-1r−/− mice received intracerebroventricular saline or Ex4 (1.0 μg). Food intake (C) and body weight change (D) were measured over 24 h. Data are represented as means ± SE. *P < 0.05 vs. saline.

Close modal

Because Ex4 (14), but not the DPP-4 inhibitor sitagliptin (16), produces weight loss in patients, it is critical that we better understand the unique anorectic properties of Ex4. To this end, we report key distinctions between the central anorectic effects of Ex4 and native GLP-1. Not only do our data confirm that central GLP-1 and Ex4 differ significantly in potency and duration of action, but they also reveal novel differences between the two peptides regarding sensitivity to GLP-1r antagonism.

Ex4, when administered into the CNS, reduces food intake in a manner distinct from that of GLP-1. Consistent with previous reports (32,33), central Ex4 reduced 4-h food intake at doses 30- to 100-fold lower than those required by GLP-1 to cause equivalent anorexia. Importantly, this difference in potency at 4 h cannot simply be explained by differences in duration of action, as both 3.0 nmol of GLP-1 and 0.03 nmol of Ex4 reduced food intake to a comparable extent from 0 to 2 h and 2 to 4 h. However, in contrast to GLP-1, Ex4 dynamically reduced food intake over 24 h of observation, indicating that even at significantly lower doses, central Ex4 exhibits a significantly longer duration of action.

Consistent with our food intake data, 0.1 μg of central Ex4 produced an almost identical degree of neuronal activation as 10.0 μg of GLP-1 in the PVN and the NTS but interestingly not in the CeA. It is possible that because 100-fold less Ex4 than GLP-1 was administered, less peptide diffused through the neuropil to the CeA, which, unlike the PVN and the NTS, does not abut the ventricular system. Despite producing less neuronal activation in the CeA, an area important for the formation of GLP-1–mediated CTA (34), central Ex4 produced a lower preference ratio for saccharin than GLP-1, suggesting that Ex4 induced a greater visceral illness response. Although a proportional relationship between GLP-1r–mediated neuronal activity and behavioral responses has yet to be established, these data are significant because they suggest that enhanced visceral illness or aversive learning may in part underlie the potent anorectic effect of central Ex4.

The above data and those of others support a role for Ex4 as a highly potent, long-acting CNS GLP-1r agonist, yet the mechanism for this unique pharmacological profile remains unknown. One possibility is that GLP-1 and Ex4 bind differently to CNS GLP-1r. However, in vitro and ex vivo comparisons of binding affinity have yielded equivocal results (35,,38), and it remains unclear whether GLP-1 and Ex4 remain bound to CNS GLP-1r for different periods of time. A second possibility is that GLP-1 and Ex4 differentially desensitize CNS GLP-1r. However, data from Baggio et al. (39) revealed no difference in the ability of GLP-1 and Ex4 to desensitize the GLP-1r in vitro. Moreover, because GLP-1 and Ex4 were administered as boluses, this hypothesis fails to adequately explain the present results. Finally, it is possible that differential clearance and/or degradation of GLP-1 versus Ex4 account for their distinct pharmacological profiles within the CNS.

Perhaps the most striking difference between central GLP-1 and Ex4 revealed by our data are their sensitivity to GLP-1r antagonism. Whereas dHEx almost completely blocked anorexia and neuronal activation induced by GLP-1, it failed to significantly block that induced by an equipotent dose of Ex4. This phenomenon is not specific to dHEx, as Ex9 also failed to block anorexia induced by central Ex4. However, dHEx is an effective antagonist of Ex4 in vitro, as it completely blocked the enhancement of glucose-stimulated insulin secretion induced by Ex4 in INS-1 cells. In addition, dHEx is an effective antagonist of Ex4 in vivo, as intraperitoneal dHEx completely blocked anorexia induced by intraperitoneal Ex4. Taken together, these data indicate that compared with GLP-1, Ex4 is relatively insensitive to GLP-1r antagonism. Moreover, this phenomenon seems to be specific for central effects but not peripheral effects, many of which have been reported to be blocked by Ex9 (40,42).

Certainly, previous studies (22,24) have reported an inability to block Ex4 effects with GLP-1r antagonists. However, they may not have adequately accounted for the increased potency of Ex4 versus GLP-1. Here, we closely controlled for this difference and found that pretreatment with GLP-1r antagonists significantly blocked anorexia induced by central GLP-1 but not an equipotent and, importantly, 100-fold lower dose of Ex4. Moreover, this phenomenon is not secondary to differences in agonist duration of action, as it was observed at early time points when both GLP-1 and Ex4 dynamically reduced food intake. Nor is it secondary to the antagonist duration of action, as timing of pretreatment and treatment injections was consistent across all experiments, and similar trends were observed with both c-Fos and food intake (data not shown) at 2 h.

While intriguing, these data are difficult to reconcile with our other experiments. Specifically, we found no differences between GLP-1 and Ex4 in either potency or sensitivity to dHEx in vitro, although this discrepancy might easily be explained by obvious differences between animal models and immortalized cell lines. More difficult to explain, however, is the comparison to our peripheral Ex4 food intake study, in which the same 100-fold excess of dHEx, this time administered intraperitoneally, completely blocked intraperitoneal Ex4-induced anorexia. Consequently, it is possible that fundamental differences exist between central and peripheral GLP-1r, which may occur at the level of posttranslational processing, protein–protein interactions, or coupling to second-messenger systems.

Perhaps the most obvious explanation for the discrepancies between central GLP-1 and Ex4 is that the latter acts in part independently of the GLP-1r. However, consistent with previous reports (18), central Ex4 had no effect on either food intake or body weight in GLP-1r−/− mice, suggesting that the GLP-1r is required for these effects. Although the lack of Ex4 effects in GLP-1r−/− mice provides a strong basis to rule out GLP-1r independence, there is some evidence for both functional (43) and structural (44) differences between the GLP-1 systems of mice and rats. Recently, Sowden et al. (20) reported that Ex4 increases heart rate in rats but not mice. Finally, other studies reporting an inability to block Ex4 effects with GLP-1r antagonists have all been conducted in rats (22,24). Although none of these observations provides definitive evidence for GLP-1r–independent effects of Ex4, they do raise the possibility that Ex4 may interact with the GLP-1r in a species-dependent manner.

Although difficult to reconcile with the above data, our findings regarding central Ex4 and GLP-1r antagonists are consistent with several reports of in vivo effects of Ex4 that are insensitive to GLP-1r antagonists (22,24). Recently, it was reported that central Ex4 decreases ghrelin secretion in fasted rats (24). Not only was this effect insensitive to Ex9 blockade, it was also elicited by Ex9 alone, consistent with several in vitro reports of independent Ex9 effects (45,,,,50). Whereas these data, like ours, fail to prove GLP-1r independence of Ex4, they are nonetheless significant in that they provide potential mechanistic insight into the unique anorectic properties of central Ex4, particularly its duration of action. For instance, ongoing GLP-1r signaling by Ex4 may prevent circulating ghrelin levels from rising in response to Ex4-mediated reductions in food intake, leading to an attenuation or delay in the subsequent drive to eat and thus a prolonged duration of anorexia. However, because our experiments used ad libitum–fed rats, whose circulating ghrelin levels should be low, and because Ex4 is more efficacious in fed versus fasted rats (51), it seems unlikely that Ex4's effects on ghrelin secretion underlie either its increased potency acutely or its insensitivity to GLP-1r antagonists in the present studies.

Because studies have generally found no effect of Ex4 in GLP-1r−/− mice (17,,,21), it seems reasonable to cite strictly pharmacological differences when explaining discrepancies between in vivo effects of GLP-1 and Ex4. However, in many ways, the existing data fail to adequately support this hypothesis. For instance, some in vitro studies have found Ex4 to have greater potency and affinity for the GLP-1r than native GLP-1 (35), but these differences, at least in potency, are significantly smaller than those reported here. Regarding antagonist sensitivity, one potential explanation for our findings is that Ex4 is more able to displace antagonists from the GLP-1r. However, studies have generally reported little to no difference in the ability of GLP-1 versus Ex4 to displace radio-labeled Ex9 (52,,55). Taken together, our data, combined with the existing literature, provide conclusive evidence for distinct pharmacological profiles of GLP-1 and Ex4, yet further studies are needed to understand whether pharmacological differences alone are sufficient to explain the unique in vivo effects of Ex4.

In conclusion, our data indicate that the central, but not peripheral, anorectic effect of Ex4 is insensitive to GLP-1r antagonism, yet GLP-1r is required for this effect. These data suggest that there are important differences between the in vivo pharmacological properties of GLP-1 and Ex4 within the CNS. Moreover, they underscore the need for a greater understanding of how these ligands interact with CNS GLP-1r, particularly in light of recent data revealing novel roles of CNS GLP-1r activity in the regulation of peripheral glucose homeostasis and cardiovascular function (56,57). Such an understanding is critical if we are to maximize the therapeutic benefit of Ex4 and other GLP-1–based therapies.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

This work was supported by National Institutes of Health (NIH) Grant RO1 DK54890 (to R.J.S.), American Diabetes Association Physician Scientist Training Award 7-06-PST-02 (to J.G.B.), NIH Grant RO1 DK57900 (to D.A.D.), Juvenile Diabetes Research Foundation Grant 1-2006-796 (to D.J.D.), and research funding from Amylin Pharmaceuticals.

D.J.D. has served as an advisor or consultant to Amylin Pharmaceuticals, Arena Pharmaceuticals, Arisaph Pharmaceuticals, Conjuchem, Eli Lilly, Emisphere Technologies, GlaxoSmithKline, Glenmark Pharmaceuticals, Hoffman LaRoche, Isis Pharmaceuticals, MannKind, Merck Research Laboratories, Metabolex, and Novartis Pharmaceuticals. Neither D.J.D. nor his family members hold stock directly or indirectly in any of these companies. No other potential conflicts of interest relevant to this article were reported.

We thank Kathleen Smith, Joyce Sorrell, and Jeanette Teague for their expert technical assistance.

1.
Lopez
LC
,
Frazier
ML
,
Su
CJ
,
Kumar
A
,
Saunders
GF
:
Mammalian pancreatic preproglucagon contains three glucagon-related peptides
.
Proc Natl Acad Sci U S A
1983
; 
80
:
5485
5489
2.
Varndell
IM
,
Bishop
AE
,
Sikri
KL
,
Uttenthal
LO
,
Bloom
SR
,
Polak
JM
:
Localization of glucagon-like peptide (GLP) immunoreactants in human gut and pancreas using light and electron microscopic immunocytochemistry
.
J Histochem Cytochem
1985
; 
33
:
1080
1086
3.
Merchenthaler
I
,
Lane
M
,
Shughrue
P
:
Distribution of pre-pro-glucagon and glucagon-like peptide-1 receptor messenger RNAs in the rat central nervous system
.
J Comp Neurol
1999
; 
403
:
261
280
4.
Shimizu
I
,
Hirota
M
,
Ohboshi
C
,
Shima
K
:
Identification and localization of glucagon-like peptide-1 and its receptor in the brain
.
Endocrinology
1987
; 
121
:
1076
1082
5.
Turton
MD
,
O'Shea
D
,
Gunn
I
,
Beak
SA
,
Edwards
CM
,
Meeran
K
,
Choi
SJ
,
Taylor
GM
,
Heath
MM
,
Lambert
PD
,
Wilding
JP
,
Smith
DM
,
Ghatei
MA
,
Herbert
J
,
Bloom
SR
:
A role for glucagon-like peptide-1 in the central regulation of feeding
.
Nature
1996
; 
379
:
69
72
[
see comments
]
6.
Tang-Christensen
M
,
Larsen
PJ
,
Goke
R
,
Fink-Jensen
A
,
Jessop
DS
,
Moller
M
,
Sheikh
SP
:
Central administration of GLP-1-(7-36) amide inhibits food and water intake in rats
.
Am J Physiol
1996
; 
271
:
R848
R856
7.
Meeran
K
,
O'Shea
D
,
Edwards
CM
,
Turton
MD
,
Heath
MM
,
Gunn
I
,
Abusnana
S
,
Rossi
M
,
Small
CJ
,
Goldstone
AP
,
Taylor
GM
,
Sunter
D
,
Steere
J
,
Choi
SJ
,
Ghatei
MA
,
Bloom
SR
:
Repeated intracerebroventricular administration of glucagon-like peptide-1-(7-36) amide or exendin-(9-39) alters body weight in the rat
.
Endocrinology
1999
; 
140
:
244
250
8.
Kreymann
B
,
Ghatei
MA
,
Williams
G
,
Bloom
SR
:
Glucagon-like peptide-1 7-36: a physiological incretin in man
.
Lancet
1987
; 
2
:
1300
1303
9.
Mentlein
R
,
Gallwitz
B
,
Schmidt
WE
:
Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon-like peptide-1 (7-36) amide, petide histidine methionine and is responsible for their degradation in human serum
.
Eur J Biochem
1993
; 
214
:
829
835
10.
Deacon
CF
,
Nauck
MA
,
Toft-Nielsen
M
,
Pridal
L
,
Willms
B
,
Holst
JJ
:
Both subcutaneously and intravenously administered glucagon-like peptide I are rapidly degraded from the NH2-terminus in type II diabetic patients and in healthy subjects
.
Diabetes
1995
; 
44
:
1126
1131
11.
Kieffer
TJ
,
McIntosh
CH
,
Pederson
RA
:
Degradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IV
.
Endocrinology
1995
; 
136
:
3585
3596
12.
Young
AA
,
Gedulin
BR
,
Bhavsar
S
,
Bodkin
N
,
Jodka
C
,
Hansen
B
,
Denaro
M
:
Glucose-lowering and insulin-sensitizing actions of exendin-4: studies in obese diabetic (ob/ob, db/db) mice, diabetic fatty Zucker rats, and diabetic rhesus monkeys (Macaca mulatta)
.
Diabetes
1999
; 
48
:
1026
1034
13.
Greig
NH
,
Holloway
HW
,
De Ore
KA
,
Jani
D
,
Wang
Y
,
Zhou
J
,
Garant
MJ
,
Egan
JM
:
Once daily injection of exendin-4 to diabetic mice achieves long-term beneficial effects on blood glucose concentrations
.
Diabetologia
1999
; 
42
:
45
50
14.
Kendall
DM
,
Riddle
MC
,
Rosenstock
J
,
Zhuang
D
,
Kim
DD
,
Fineman
MS
,
Baron
AD
:
Effects of exenatide (exendin-4) on glycemic control over 30 weeks in patients with type 2 diabetes treated with metformin and a sulfonylurea
.
Diabetes Care
2005
; 
28
:
1083
1091
15.
Herman
GA
,
Bergman
A
,
Stevens
C
,
Kotey
P
,
Yi
B
,
Zhao
P
,
Dietrich
B
,
Golor
G
,
Schrodter
A
,
Keymeulen
B
,
Lasseter
KC
,
Kipnes
MS
,
Snyder
K
,
Hilliard
D
,
Tanen
M
,
Cilissen
C
,
De Smet
M
,
de Lepeleire
I
,
Van Dyck
K
,
Wang
AQ
,
Zeng
W
,
Davies
MJ
,
Tanaka
W
,
Holst
JJ
,
Deacon
CF
,
Gottesdiener
KM
,
Wagner
JA
:
Effect of single oral doses of sitagliptin, a dipeptidyl peptidase-4 inhibitor, on incretin and plasma glucose levels after an oral glucose tolerance test in patients with type 2 diabetes
.
J Clin Endocrinol Metab
2006
; 
91
:
4612
4619
16.
Raz
I
,
Hanefeld
M
,
Xu
L
,
Caria
C
,
Williams-Herman
D
,
Khatami
H
:
Efficacy and safety of the dipeptidyl peptidase-4 inhibitor sitagliptin as monotherapy in patients with type 2 diabetes mellitus
.
Diabetologia
2006
; 
49
:
2564
2571
17.
Hansotia
T
,
Baggio
LL
,
Delmeire
D
,
Hinke
SA
,
Yamada
Y
,
Tsukiyama
K
,
Seino
Y
,
Holst
JJ
,
Schuit
F
,
Drucker
DJ
:
Double incretin receptor knockout (DIRKO) mice reveal an essential role for the enteroinsular axis in transducing the glucoregulatory actions of DPP-IV inhibitors
.
Diabetes
2004
; 
53
:
1326
1335
18.
Baggio
LL
,
Huang
Q
,
Brown
TJ
,
Drucker
DJ
:
Oxyntomodulin and glucagon-like peptide-1 differentially regulate murine food intake and energy expenditure
.
Gastroenterology
2004
; 
127
:
546
558
19.
Baggio
LL
,
Huang
Q
,
Brown
TJ
,
Drucker
DJ
:
A recombinant human glucagon-like peptide (GLP)-1-albumin protein (albugon) mimics peptidergic activation of GLP-1 receptor-dependent pathways coupled with satiety, gastrointestinal motility, and glucose homeostasis
.
Diabetes
2004
; 
53
:
2492
2500
20.
Sowden
GL
,
Drucker
DJ
,
Weinshenker
D
,
Swoap
SJ
:
Oxyntomodulin increases intrinsic heart rate in mice independent of the glucagon-like peptide-1 receptor
.
Am J Physiol Regul Integr Comp Physiol
2007
; 
292
:
R962
R970
21.
Baggio
LL
,
Huang
Q
,
Cao
X
,
Drucker
DJ
:
An albumin-exendin-4 conjugate engages central and peripheral circuits regulating murine energy and glucose homeostasis
.
Gastroenterology
2008
; 
134
:
1137
1147
22.
Malendowicz
LK
,
Nowak
KW
:
Preproglucagon derived peptides and thyrotropin (TSH) secretion in the rat: robust and sustained lowering of blood TSH levels in exendin-4 injected animals
.
Int J Mol Med
2002
; 
10
:
327
331
23.
Malendowicz
LK
,
Nussdorfer
GG
,
Nowak
KW
,
Ziolkowska
A
,
Tortorella
C
,
Trejter
M
:
Exendin-4, a GLP-1 receptor agonist, stimulates pituitary-adrenocortical axis in the rat: investigations into the mechanism(s) underlying Ex4 effect
.
Int J Mol Med
2003
12
237
241
24.
Perez-Tilve
D
,
Gonzalez-Matias
L
,
Alvarez-Crespo
M
,
Leiras
R
,
Tovar
S
,
Dieguez
C
,
Mallo
F
:
Exendin-4 potently decreases ghrelin levels in fasting rats
.
Diabetes
2007
; 
56
:
143
151
25.
Chavez
M
,
Kaiyala
K
,
Madden
LJ
,
Schwartz
MW
,
Woods
SC
:
Intraventricular insulin and the level of maintained body weight in rats
.
Behav Neurosci
1995
; 
109
:
528
531
26.
Brown
LM
,
Clegg
DJ
,
Benoit
SC
,
Woods
SC
:
Intraventricular insulin and leptin reduce food intake and body weight in C57BL/6J mice
.
Physiol Behav
2006
; 
89
:
687
691
27.
Cota
D
,
Proulx
K
,
Smith
KA
,
Kozma
SC
,
Thomas
G
,
Woods
SC
,
Seeley
RJ
:
Hypothalamic mTOR signaling regulates food intake
.
Science
2006
; 
312
:
927
930
28.
Montrose-Rafizadeh
C
,
Yang
H
,
Rodgers
BD
,
Beday
A
,
Pritchette
LA
,
Eng
J
:
High potency antagonists of the pancreatic glucagon-like peptide-1 receptor
.
J Biol Chem
1997
; 
272
:
21201
21206
29.
D'Alessio
DA
,
Fujimoto
WY
,
Ensinck
JW
:
Effects of glucagonlike peptide I-(7-36) on release of insulin, glucagon, and somatostatin by rat pancreatic islet cell monolayer cultures
.
Diabetes
1989
; 
38
:
1534
1538
30.
Seeley
RJ
,
Blake
K
,
Rushing
PA
,
Benoit
SC
,
Eng
J
,
Woods
SC
,
D'Alessio
D
:
The role of CNS GLP-1-(7-36) amide receptors in mediating the visceral illness effects of lithium chloride
.
J Neurosci
2000
; 
20
:
1616
1621
31.
Yamamoto
H
,
Lee
CE
,
Marcus
JN
,
Williams
TD
,
Overton
JM
,
Lopez
ME
,
Hollenberg
AN
,
Baggio
L
,
Saper
CB
,
Drucker
DJ
,
Elmquist
JK
:
Glucagon-like peptide-1 receptor stimulation increases blood pressure and heart rate and activates autonomic regulatory neurons
.
J Clin Invest
2002
; 
110
:
43
52
32.
Rodriquez de Fonseca
F
,
Navarro
M
,
Alvarez
E
,
Roncero
I
,
Chowen
JA
,
Maestre
O
,
Gomez
R
,
Munoz
RM
,
Eng
J
,
Blazquez
E
:
Peripheral versus central effects of glucagon-like peptide-1 receptor agonists on satiety and body weight loss in Zucker obese rats
.
Metabolism
2000
; 
49
:
709
717
33.
Navarro
M
,
Rodriquez de Fonseca
F
,
Alvarez
E
,
Chowen
JA
,
Zueco
JA
,
Gomez
R
,
Eng
J
,
Blazquez
E
:
Colocalization of glucagon-like peptide-1 (GLP-1) receptors, glucose transporter GLUT-2, and glucokinase mRNAs in rat hypothalamic cells: evidence for a role of GLP-1 receptor agonists as an inhibitory signal for food and water intake
.
J Neurochem
1996
; 
67
:
1982
1991
34.
Kinzig
KP
,
D'Alessio
DA
,
Seeley
RJ
:
The diverse roles of specific GLP-1 receptors in the control of food intake and the response to visceral illness
.
J Neurosci
2002
; 
22
:
10470
10476
35.
Goke
R
,
Fehmann
HC
,
Linn
T
,
Schmidt
H
,
Krause
M
,
Eng
J
,
Goke
B
:
Exendin-4 is a high potency agonist and truncated exendin-(9-39)-amide an antagonist at the glucagon-like peptide 1-(7-36)-amide receptor of insulin-secreting beta-cells
.
J Biol Chem
1993
; 
268
:
19650
19655
36.
Fehmann
HC
,
Jiang
J
,
Schweinfurth
J
,
Wheeler
MB
,
Boyd
AE
 3rd
,
Goke
B
:
Stable expression of the rat GLP-I receptor in CHO cells: activation and binding characteristics utilizing GLP-I(7-36)-amide, oxyntomodulin, exendin-4, and exendin(9-39)
.
Peptides
1994
; 
15
:
453
456
37.
Goke
R
,
Larsen
PJ
,
Mikkelsen
JD
,
Sheikh
SP
:
Identification of specific binding sites for glucagon-like peptide-1 on the posterior lobe of the rat pituitary
.
Neuroendocrinology
1995
; 
62
:
130
134
38.
Goke
R
,
Larsen
PJ
,
Mikkelsen
JD
,
Sheikh
SP
:
Distribution of GLP-1 binding sites in the rat brain: evidence that exendin-4 is a ligand of brain GLP-1 binding sites
.
Eur J Neurosci
1995
; 
7
:
2294
2300
39.
Baggio
LL
,
Kim
J-G
,
Drucker
DJ
:
Chronic exposure to GLP-1r agonists promotes homologous GLP-1 receptor desensitization in vitro but does not attenuate GLP-1r–dependent glucose homeostasis in vivo
.
Diabetes
2004
; 
53
(
Suppl. 3
):
S205
S214
40.
Kolligs
F
,
Fehmann
HC
,
Goke
R
,
Goke
B
:
Reduction of the incretin effect in rats by the glucagon-like peptide 1 receptor antagonist exendin (9-39) amide
.
Diabetes
1995
; 
44
:
16
19
41.
Barragan
JM
,
Rodriguez
RE
,
Eng
J
,
Blazquez
E
:
Interactions of exendin (9-39) with the effects of GLP-1 (7-36) amide and of exendin-4 on arterial blood pressure and heart rate in rats
.
Reg Peptides
1996
; 
67
:
63
68
42.
Benito
E
,
Blazquez
E
,
Bosch
MA
:
Glucagon-like peptide-1-(7-36)amide increases pulmonary surfactant secretion through a cyclic adenosine 3′, 5′-monophosphate-dependent protein kinase mechanism in rat type II pneumocytes
.
Endocrinology
1998
; 
139
:
2363
2368
43.
Lachey
JL
,
D'Alessio
DA
,
Rinaman
L
,
Elmquist
JK
,
Drucker
DJ
,
Seeley
RJ
:
The role of central glucagon-like peptide-1 in mediating the effects of visceral illness: differential effects in rats and mice
.
Endocrinology
2005
; 
146
:
458
462
44.
Huo
L
,
Gamber
KM
,
Grill
HJ
,
Bjorbaek
C
:
Divergent leptin signaling in proglucagon neurons of the nucleus of the solitary tract in mice and rats
.
Endocrinology
2008
; 
149
:
492
497
45.
Montrose-Rafizadeh
C
,
Yang
H
,
Wang
Y
,
Roth
J
,
Montrose
MH
,
Adams
LG
:
Novel signal transduction and peptide specificity of glucagon-like peptide receptor in 3T3-L1 adipocytes
.
J Cell Physiol
1997
; 
172
:
275
283
46.
Yang
H
,
Egan
JM
,
Wang
Y
,
Moyes
CD
,
Roth
J
,
Montrose
MH
,
Montrose-Rafizadeh
C
:
GLP-1 action in L6 myotubes is via a receptor different from the pancreatic GLP-1 receptor
.
Am J Physiol
1998
; 
275
:
C675
C683
47.
Sancho
V
,
Trigo
MV
,
Gonzalez
N
,
Valverde
I
,
Malaisse
WJ
,
Villanueva-Penacarrillo
ML
:
Effects of glucagon-like peptide-1 and exendins on kinase activity, glucose transport and lipid metabolism in adipocytes from normal and type-2 diabetic rats
.
J Mol Endocrinol
2005
; 
35
:
27
38
48.
Sancho
V
,
Nuche
B
,
Arnes
L
,
Cancelas
J
,
Gonzalez
N
,
Diaz-Miguel
M
,
Martin-Duce
A
,
Valverde
I
,
Villanueva-Penacarrillo
ML
:
The action of GLP-1 and exendins upon glucose transport in normal human adipocytes, and on kinase activity as compared to morbidly obese patients
.
Int J Mol Med
2007
; 
19
:
961
966
49.
Gonzalez
N
,
Acitores
A
,
Sancho
V
,
Valverde
I
,
Villanueva-Penacarrillo
ML
:
Effect of GLP-1 on glucose transport and its cell signalling in human myocytes
.
Regul Pept
2005
; 
126
:
203
211
50.
Arnes
L
,
Gonzalez
N
,
Tornero-Esteban
P
,
Sancho
V
,
Acitores
A
,
Valverde
I
,
Delgado
E
,
Villanueva-Penacarrillo
ML
:
Characteristics of GLP-1 and exendins action upon glucose transport and metabolism in type 2 diabetic rat skeletal muscle
.
Int J Mol Med
2008
; 
22
:
127
132
51.
Williams
DL
,
Baskin
DG
,
Schwartz
MW
:
Leptin regulation of the anorexic response to glucagon-like peptide-1 receptor stimulation
.
Diabetes
2006
; 
55
:
3387
3393
52.
Lopez de Maturana
R
,
Donnelly
D
:
The glucagon-like peptide-1 receptor binding site for the N-terminus of GLP-1 requires polarity at Asp198 rather than negative charge
.
FEBS Lett
2002
; 
530
:
244
248
53.
Lopez de Maturana
R
,
Willshaw
A
,
Kuntzsch
A
,
Rudolph
R
,
Donnelly
D
:
The isolated N-terminal domain of the glucagon-like peptide-1 (GLP-1) receptor binds exendin peptides with much higher affinity than GLP-1
.
J Biol Chem
2003
; 
278
:
10195
10200
54.
Al-Sabah
S
,
Donnelly
D
:
A model for receptor-peptide binding at the glucagon-like peptide-1 (GLP-1) receptor through the analysis of truncated ligands and receptors
.
Br J Pharmacol
2003
; 
140
:
339
346
55.
Runge
S
,
Schimmer
S
,
Oschmann
J
,
Schiodt
CB
,
Knudsen
SM
,
Jeppesen
CB
,
Madsen
K
,
Lau
J
,
Thogersen
H
,
Rudolph
R
:
Differential structural properties of GLP-1 and exendin-4 determine their relative affinity for the GLP-1 receptor N-terminal extracellular domain
.
Biochemistry
2007
; 
46
:
5830
5840
56.
Cabou
C
,
Campistron
G
,
Marsollier
N
,
Leloup
C
,
Cruciani-Guglielmacci
C
,
Penicaud
L
,
Drucker
DJ
,
Magnan
C
,
Burcelin
R
:
Brain glucagon-like peptide-1 regulates arterial blood flow, heart rate, and insulin sensitivity
.
Diabetes
2008
; 
57
:
2577
2587
57.
Knauf
C
,
Cani
PD
,
Ait-Belgnaoui
A
,
Benani
A
,
Dray
C
,
Cabou
C
,
Colom
A
,
Uldry
M
,
Rastrelli
S
,
Sabatier
E
,
Godet
N
,
Waget
A
,
Penicaud
L
,
Valet
P
,
Burcelin
R
:
Brain glucagon-like peptide 1 signaling controls the onset of high-fat diet-induced insulin resistance and reduces energy expenditure
.
Endocrinology
2008
; 
149
:
4768
4777
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.