Next Article in Journal
Wearable Activity Trackers to Improve Physical Activity and Cardiovascular Risk in Type 2 Diabetes: A Randomized Pilot Study
Previous Article in Journal
Diabetes-Induced Osteoporosis: Dual Energy X-Ray Absorptiometry Bone Quality Is Better than Bone Quantity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Liraglutide Increases Gastric Fundus Tonus and Reduces Food Intake in Type 2 Diabetic Rats

by
Ana Catarina Carrêlo
1,2,3,
Beatriz Martins
1,2,4,
Raquel Seiça
3,
Carlos Fontes-Ribeiro
4,
Paulo Matafome
2,3,5,6,* and
Sónia Silva
1,2,5,*
1
Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
2
Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
3
Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
4
Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
5
Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
6
Coimbra Health School (ESTeSC), Health & Technology Research Center (H&TRC), Polytechnic University of Coimbra, 3046-854 Coimbra, Portugal
*
Authors to whom correspondence should be addressed.
Diabetology 2025, 6(9), 96; https://doi.org/10.3390/diabetology6090096
Submission received: 3 June 2025 / Revised: 31 July 2025 / Accepted: 25 August 2025 / Published: 5 September 2025

Abstract

Background/Objectives: Incretin-based therapies have demonstrated benefits in glycemic control and the prevention of long-term complications of diabetes. In addition to glucose-dependent insulin secretion stimulation, glucagon-like peptide-1 (GLP-1) also inhibits gastric acid secretion, delays gastric emptying, inhibits gut motility and induces satiety. We aimed to understand the modulation of gastric fundus motility by GLP-1 receptor agonists (GLP-1RA). Methods: We have studied the relaxation to sodium nitroprusside (SNP) and noradrenaline (NA) of gastric fundus isolated from Wistar rats and Goto-Kakizaki (GK) rats, an animal model of spontaneous non-obese type 2 diabetes, after Liraglutide treatment (200 μg/kg s.c., b.i.d., 14 days). Results: Decreased relaxation induced by SNP and NA (0.01–889 μM) was observed in treated groups, with no significant changes in SNP maximum relaxation or in nNOS/p-nNOS levels between treated and non-treated rats of both animal models. Accordingly, in rat gastric fundus pre-contracted with 5 µM of carbachol, GLP-1RA (0.05–111.1 nM) induced contractile responses that were GLP-1R-dependent and -independent. Exenatide showed more intrinsic activity, while Liraglutide showed more potency than GLP-1 in Wistar rats. Moreover, GLP-1 showed more intrinsic activity in diabetic rats compared to control ones. Conclusions: Liraglutide-induced increased gastric muscle tone may contribute to the significant decrease in caloric intake and body weight in all treated rats, causing a reduction in gastric accommodation during food intake. Thus, the increased gastric fundus tone induced by GLP-1RA may constitute a peripheral mechanism by which they can reduce food intake and induce satiety.

1. Introduction

Type 2 Diabetes Mellitus (T2DM) is the most common type of diabetes, accounting for a higher risk of cardiovascular diseases in later life according to the last International Diabetes Federation (IDF) report [1]. Projections of the global prevalence of diabetes in the 20–79 age group will be 853 million people worldwide in 2050 [1]. T2DM is a long-term metabolic disorder with high rates of socioeconomic costs, morbidity and mortality. Person-centered pharmacological interventions, lifestyle/behavioral changes and moderate-intensity physical activity are used in preventing or delaying it and improving other cardiometabolic markers [2].
Incretin-based antidiabetic therapies, namely glucagon-like peptide (GLP)-1 receptor agonists (GLP-1RA) and dipeptidyl peptidase-4 (DPP-4) inhibitors, have demonstrated benefits in glycemic control by stimulating nutrient-induced insulin secretion while inhibiting glucagon secretion. They also prevent diabetes long-term complications [3], although the cardioprotective effects associated with the GLP-1 metabolite, GLP-1 (9–36) amide—which is inactive at the GLP-1 receptor (GLP-1R)—are absent in DPP-4-resistant GLP-1 analogs and DPP-4 inhibitors [4]. This alerted the scientific community to the possibility that GLP-1’s actions could be mediated by GLP-1R-independent and -dependent mechanisms [5]. GLP-1RA are a particularly attractive choice for T2DM treatment because these drugs preserve β-cell function and have beneficial effects on hypertension, hyperlipidemia and obesity, being recommended as a preferred treatment for patients with pre-existing atherosclerotic cardiovascular disease and microvascular diabetic complications. Moreover, they do not cause side effects observed for other antidiabetic drugs, such as hypoglycemia and weight gain [6]. On the contrary, all approved GLP-1RA have the potential to induce weight loss by decreasing appetite and increasing satiety, mainly through an interaction with GLP-1R in brain areas involved in energy homeostasis. Indeed, murine models show that liraglutide— an acylated analog of human GLP-1, sharing 97% amino acid sequence homology and exhibiting non-covalent binding to serum albumin in circulation, thereby conferring high resistance to DPP-4-mediated degradation. [3,6,7,8]—can access specific brain areas involved in appetite regulation. It binds to GLP-1R on pro-opiomelanocortin and cocaine- and amphetamine-regulated transcript (POMC/CART) expressing neurons in the arcuate nucleus, thereby increasing satiety. GLP-1RA are approved for both long-term weight management and T2DM [9] and are associated with reductions in appetite and hunger, lower preference for energy-dense foods, alteration in food reward pathways, decrease in food craving, and improvement in eating control [10].
Corroborating these findings, GLP-1RA treatment has also been shown to delay gastric emptying within the first postprandial hour, reducing food intake and post-meal rises in glycaemia. However, tachyphylaxis has been described for long-acting GLP-1RA, such as once-daily subcutaneous liraglutide, suggesting additional mechanisms of action in GLP-1RA-mediated weight loss [11,12]. In contrast, short-acting GLP-1RA, such as twice-daily subcutaneous exenatide—which shares 53% amino acid sequence identity with endogenous GLP-1 and is also resistant to DPP-4-mediated degradation—have been associated with gastrointestinal adverse effects, including nausea, vomiting, and diarrhea, which appear to be transient with long-acting ones [6,13].
Besides pancreatic islets and peripheral and central nervous systems, the GLP-1R is expressed in the kidney, lung, heart, gastrointestinal tract, adipose tissue and smooth muscle [14,15,16]. In vascular smooth muscle, GLP-1-induced activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (PKB/Akt) signaling pathway leads to phosphorylation and activation of the endothelial isoform of nitric oxide synthase (eNOS), resulting in nitric oxide (NO) release and subsequent vascular smooth muscle relaxation [17].
In the gut, GLP-1 inhibits gastric acid secretion, delays gastric emptying, as mentioned above, and inhibits gut motility through activation of GLP-1R on enteric neurons, possibly via NO release [18,19,20,21,22]. However, its effect on the gastric fundus motility was never addressed before. Given that this region is responsible for accommodating ingested food and that reducing its tone will allow an increase in gastric volume, without a substantial increase in intragastric pressure [23], we hypothesized that GLP-1RA may modulate the contractile response of the gastric fundus, reducing gastric compliance and thereby limiting the accommodation of food in the stomach. This contributes to satiety through a peripheral mechanism independent of delayed gastric emptying.
The present study aimed to understand the mechanisms involved in the modulation of gastric fundus motility by GLP-1RA, with a focus on Liraglutide, as well as the alterations induced by T2DM in such mechanisms. To this end, relaxation to sodium nitroprusside (SNP) and noradrenaline (NA) was studied in gastric fundus isolated from Wistar rats and Goto-Kakizaki (GK) rats, an animal model of spontaneous non-obese T2DM, after Liraglutide treatment. The contractile responses of gastric fundus strips, isolated from untreated animals and pre-contracted with carbachol (CCh), to native GLP-1, the long-acting GLP-1A Liraglutide, and the short-acting agonist exenatide were further evaluated.

2. Materials and Methods

2.1. Drugs and Chemicals

The following drugs were used in functional studies: GLP-1 (7–36) amide, Exenatide (Exendin-4), Exendin-3 (9–39) amide, from Tocris, Bristol, UK; Liraglutide (Victoza®) from Novo Nordisk, Bagsværd, Denmark; NA and SNP from Sigma-Aldrich, St Louis, MO, USA. Solutions of these drugs were prepared fresh on the day of the experiment with an adequate solvent.
The antibodies used in molecular studies were Calnexin (Sicgen, Coimbra, Portugal), GLP-1R (bs-1559R, Bioss, Woburn, MA, USA), nNOS and nNOS-S1417 (ab5586, ab5583, Abcam, Waltham, MA, USA). The secondary antibodies were anti-rabbit and anti-goat (Bio-Rad, Hercules, CA, USA).
Other reagents used in this study were purchased from Panreac (Barcelona, Spain) and Sigma-Aldrich (St. Louis, MO, USA).

2.2. Animals

Male Wistar and GK rats were obtained from our local breeding colonies at the Faculty of Medicine, University of Coimbra, Portugal. All animals were maintained under controlled environmental conditions (day-night cycles of 12 h, temperature of 22–24 °C and humidity of 50–60%) with ad libitum access to food (standard diet A03, SAFE®, Augy, France) and water. The experimental protocol was approved by the local Institutional Animal Care and Use Committee, and all the procedures were performed by licensed users of the Federation of Laboratory Animal Science Associations (FELASA).

2.3. In Vivo Studies

Wistar and GK rats with 14 weeks of age were blinded allocated into four groups: Wistar (n = 7) and GK (n = 4) rats treated with Liraglutide (200 μg/kg s.c.), twice daily [24], for fourteen days [25] (WL and GKL, respectively); and Wistar (n = 7) and GK (n = 4) rats treated with saline (NaCl 0.9% s.c.) during the same period (WC and GKC, respectively). All animals were maintained under controlled environmental conditions with ad libitum access to food and water during the treatment.
Fasting glycemia, triglycerides, total cholesterol and intraperitoneal insulin tolerance test (ITT) were evaluated on the first and last days of treatment in blood from the tail vein and after a 6 h fasting period. For ITT an intraperitoneal injection of insulin (250 mU/kg) was performed, and the evaluation of glycaemia was performed at 0, 15, 30 and 60 min using a glucose meter and test strips (Accu-Chek Aviva, Roche, Basel, Switzerland). The area under the curve (AUC) was calculated. Triglycerides and total cholesterol were evaluated through Accutrend Plus Meter (Roche, Basel, Switzerland). Body weight and caloric intake were evaluated daily and weekly, respectively.

2.4. Functional Studies

2.4.1. Relaxation Induced by SNP or NA of Gastric Fundus Strips Isolated from Treated and Non-Treated Rats with Liraglutide

At day 15, at the end of treatment with Liraglutide, animals were anesthetized with ketamine (75 mg/kg body weight; Pfizer Inc., New York, NY, USA)/chlorpromazine (3 mg/kg body weight; Vitória Laboratories, Amadora, Portugal) and then sacrificed by cervical displacement.
The stomach was isolated, and the fundus region was dissected and washed in ice-cold Krebs–Henseleit solution (mM: NaCl 118.67; KCl 5.36; MgSO4·7H2O 0.57; CaCl2·2H2O 1.90; KH2PO4 0.90; NaHCO3 25; glucose 11.1), pH 7.4, aerated with 5% CO2–95% O2. Four longitudinal fundal strips (approximately 15 mm) were obtained, following the methodology described by Riazi-Farzad et al. [26] and based on that firstly described by Vane [27], and suspended on stainless steel hooks under 19.6 mN of tension in 10 mL organ baths (Panlab, Barcelona, Spain) filled with aerated Krebs–Henseleit solution maintained at 37 °C. Gastric fundus strips were allowed to equilibrate for 2 h and then were submitted to isometric relaxations induced by two successive cumulative concentration-response (CR) curves for SNP and NA (0.01–889 μM) after pre-contraction with CCh 5 μM [28], using Panlab isometric transducers (Barcelona, Spain) connected to a four-channel polygraph (Polygraph 4006, Letica Scientific Instruments, Barcelona, Spain). The two CR curves were performed with a time interval of one hour to avoid tachyphylaxis.

2.4.2. Responses to GLP1-RA of Pre-Contracted Gastric Fundus Strips Isolated from Non-Treated Rats

Following the above-described procedure, gastric fundus strips isolated from non-treated Wistar (n = 4) and GK (n = 4) rats were mounted in organ baths, and after the equilibration period of 2 h, with periodic washings, isometric contractions of cumulative CR curves for GLP-1 (0.05–111.1 nM) were recorded after pre-contraction with CCh 5 μM (Figure 1). The efficacy and potency of the two drugs, exenatide and Liraglutide, were also evaluated in comparison to the endogenous agonist using the same experimental protocol. For adequate pharmacological characterization, a second cumulative CR curve of the full agonist GLP-1 was performed in the presence or absence of Exendin-3 (300 nM), a selective GLP-1R antagonist added thirty minutes before.

2.4.3. Analysis of Results

Relaxation responses to SNP and NA were expressed as a percentage of inhibition of the pre-contraction obtained with CCh. Contractile responses to GLP-1RA were expressed as a percentage of the pre-contraction obtained with CCh and were also analyzed in terms of mN of tension. Contractile responses of the GLP-1 second CR curve (performed in the presence or absence of Exendin-3) were expressed as a percentage of the maximum contraction obtained in the first CR curve of the respective strips. Considering that in each assay, gastric fundus strips pre-incubated with the appropriate solvent of each drug were used as controls, results were then expressed as a percentage of the control strips maximum response.
In both experimental protocols, the maximum contractile/relaxation response (Emax) and the pEC50 (negative logarithm of the molar concentration of agonist inducing half maximum response) were determined. Emax and pEC50 values represent, respectively, the intrinsic activity and the potency of a drug in eliciting a response. Values of pEC50 were obtained by interpolation in each CR curve in a half-logarithmic scale (percentage of maximum contraction/relaxation vs. logarithm of concentration in mol/L), using a computer program (CurveExpert 2.2 version for Windows, Hyams Development, London, UK).
Differences between NO donor or NA Emax and pEC50-values for the four animal groups (control and diabetic rats treated and non-treated with Liraglutide) and for different GLP-1RA were statistically evaluated.
Differences between dose-responses to CR curves for native GLP-1 performed in the absence or in the presence of Exendin-3 were also analyzed.

2.5. Western Blot Analysis

Gastric fundus samples isolated from the four groups of animals were cut into small pieces and were homogenized by mechanical disruption with lysis buffer (0.25 M Tris HCl, 125 mM NaCl, 1% Triton-X-100, 0.5% SDS, 1 mM EDTA, 1 mM EGTA, 20 mM NaF, 2 mM Na3VO4, 10 mM β-glycerophosphate, 2.5 mM of sodium pyrophosphate, 10 mM of PMSF and 40 µL of protease inhibitor) followed by centrifugation (14,000 rpm, 20 min, 4 °C). The supernatants were collected, and a second centrifugation was performed. Protein concentration of the lysates was determined using the BCA Protein Assay Kit (Bio-Rad, Hercules, CA, USA). Then the supernatants were mixed with Laemmli buffer (62.5 mM Tris-HCl, 10% glycerol, 2% SDS, 5% β-mercaptoethanol, 0.01% bromophenol blue). Polyacrylamide gels (SDS-PAGE) were loaded with the obtained samples, transferred to polyvinylidene fluoride (PVDF) membranes, which were washed and blocked with TBS-T 0.01% and 5% BSA, and incubated overnight with the primary antibodies followed by a 2 h-incubation with secondary antibodies. Membranes were revealed using ECL substrate in a Versadoc system (Bio-Rad, Hercules, CA, USA) and analyzed with Image Quant® (Molecular Dynamics, San Jose, CA, USA). To confirm equal protein loading, membranes were reprobed for calnexin and results were normalized against calnexin and then expressed as a percentage of the WC group protein density.

2.6. Immunohistochemistry

Gastric fundus samples isolated from animals of all experimental groups were also fixed in 4% formalin buffered at pH 6.9 and embedded in paraffin. Histological sections (3 μm thick) were deparaffinized in a BOND™ Dewax solution (Leica Biosystems, Chicago, IL, USA), rehydrated in 100% alcohol and then washed with a BOND™ Wash solution (Leica Biosystems). Antigen retrieval was performed using sodium citrate buffer 10% (v/v), at pH 6, and endogenous peroxidase quenching was performed using 3–4% (v/v) hydrogen peroxide (Novocastra, Peroxidase Block solution, Leica Biosystems).
Sections were then incubated with the primary rabbit polyclonal anti-GLP-1R and secondary antibodies diluted in suitable solutions (Leica Biosystems) and revealed with the chromogen diaminobenzidine (Mixed DAB Refine BOND™, Leica Biosystems) for 10 min. Finally, slides were counterstained with hematoxylin (Hematoxylin BOND™, Leica Systems) for 5 min, followed by diaphanization, dehydration and mounting in a DPX synthetic medium. Immunostaining analysis was performed using an optic microscope Nikon Eclipse 80i (Nikon, Tokyo, Japan). Histological sections were also stained with standard hematoxylin/eosin for microanatomy analysis.

2.7. Statistical Analysis

Results are presented as mean ± standard error of the mean (S.E.M.) of the number (n) of experiments indicated. Given the relatively small sample size for biological and biochemical studies (n = 4–7 animals per group) and for functional studies [s (number of gastric fundus strips tested) = 5–16 of n = 3–4 animals per group], the non-parametric Kruskal–Wallis test (all pairwise multiple comparation) was applied to determine all statistical differences between more than two groups and statistical differences between two groups were assessed using the non-parametric Mann–Whitney test for unpaired data and the Wilcoxon signed-rank test for paired data using the IBM SPSS 29 software (IBM, Armonk, NY, USA) and GraphPad Prism 9 PC Software (Boston, MA, USA). The alpha level of significance for all experiments was 0.05 and p < 0.05 was considered as the criterion for significance.

3. Results

3.1. Liraglutide Improves Metabolic Profile in Diabetic GK Rats

Liraglutide treatment significantly reduced (p < 0.05) caloric intake in Wistar and GK rats (Figure 2B), resulting in body weight loss as demonstrated by a significant reduction in body weight gain during the treatment period (WC = 0.2% ± 0.5 vs. WL = −9.6% ± 0.7, p < 0.01 and GKC = 1.2% ± 0.6 vs. GKL = −5.4% ± 2.3; p < 0.05) (Figure 2A). Regarding the analyzed biochemical parameters, comparison of initial and final values within the same animals using the Wilcoxon signed-rank test for paired data (for which § p < 0.05 means significance) revealed that Liraglutide treatment in GK rats (GKL group) led to a significant reduction only in triglyceride levels (GKL Final = 111.3. ± 6.6 vs. GKL Initial = 161.8 ± 18.4, n = 4) (Figure 2E). Additionally, we observed potential off-target effects in Wistar control and Liraglutide-treated animals with respect to the AUC during ITT (Figure 2D).
When comparing the four groups at the same time point using the Kruskal–Wallis test, the Wistar control group was used as the main reference. In this context, at baseline (Initial), GK rats exhibited significantly elevated fasting blood glucose (p < 0.01) and triglyceride levels (p < 0.05) compared to Wistar control rats at the same time point (Figure 2C and 2E, respectively). Following treatment with Liraglutide (GKL group), both fasting glycemia and triglyceride concentrations were reduced, as demonstrated by the lower statistical significance of the differences in fasting glycemia (p < 0.05), and the already mentioned significant reduction in triglyceride levels by paired data analysis. Furthermore, in untreated diabetic rats (GKC group), we observed a trend toward a worsening lipid profile (triglycerides and cholesterol) over time (Figure 2E,F, respectively), which, in contrast, may further support the beneficial effects of Liraglutide observed in the GKL group. Additionally, at baseline, GK rats exhibited reduced insulin sensitivity, as indicated by a significantly higher AUC during ITT compared to Wistar control rats at the same time point (GKL = 191.3 ± 34.1 vs. WC = 84.21 ± 2.6, p < 0.05); this difference was no longer observed following Liraglutide treatment (Final) (GKL = 143.1 ± 7.9 vs. WC = 75.41 ± 1.9, p > 0.05) (Figure 2D). Although this may suggest an improvement in insulin sensitivity in the GKL group, no definitive conclusions can be drawn based solely on the paired data analysis.
Liraglutide also altered the levels of GLP-1 receptor in the gastric fundus. A significant decrease in GLP-1R protein levels was observed in gastric fundus samples isolated from Wistar (p < 0.05) and GK (p < 0.05) rats treated with Liraglutide compared to non-treated ones. No significant changes were observed between control and diabetic rats (Figure 3A). GLP-1 receptor-positive cells were found in ganglion and capillary endothelial cells, but not on smooth muscle cells of muscularis mucosa and endothelial cells of arterioles (Figure 3B and Figure S1).

3.2. Liraglutide Treatment Reduces NO- and NA-Induced Relaxation of Gastric Fundus from Wistar and Diabetic GK Rats

SNP and NA induced smooth muscle relaxation in rat gastric fundus strips of both Wistar and GK rats in a concentration-dependent manner. Unexpectedly, Liraglutide treatment reduced the relaxation response induced by both substances when compared to the non-treated groups (Figure 4A,B). In SNP-induced relaxation, no statistically significant changes were observed in the maximum response (Emax) elicited by this NO donor between the four experimental groups in a multiple comparison test; however, a significant reduction in its potency (pEC50) was observed in Liraglutide-treated diabetic rats (GKL) compared to non-treated Wistar and GK rats (Table 1). On the other hand, a significant decrease in maximum relaxation response induced by NA was observed in Wistar-treated rats compared to non-treated ones (p < 0.005), but not between GK-treated and non-treated rats (Table 1). Furthermore, NA showed less potency in inducing relaxation of gastric fundus strips from Wistar and GK rats treated with Liraglutide (Table 1). Regarding levels of nNOS and its phosphorylated form (p-nNOS), no significant changes were observed between the gastric fundus of treated and non-treated rats (in both animal models) (Figure 4C).

3.3. GLP-1 Receptor Agonists Induce Distinct Contractile Responses of Rat Gastric Fundus Through GLP-1R-Dependent and -Independent Pathways

Due to the results obtained with the relaxation of the gastric fundus induced by a NO donor and NA, we decided to assess whether GLP-1RA would induce contraction under the same experimental conditions. Also unexpectedly, GLP-1, Exenatide and Liraglutide, caused tonic contractions of gastric fundus strips from both animal models pre-contracted with CCh 5 μM in a concentration-dependent (0.05–111.1 nM) manner (Table 2). The Exenatide showed more intrinsic activity (Emax) and Liraglutide more potency (pEC50) than GLP-1 in Wistar rats. Moreover, in GK rats, GLP-1 showed more intrinsic activity (Emax) than Liraglutide, and comparing both animal models, GLP-1 showed more intrinsic activity (Emax) in diabetic rats than in control ones (Table 2). Accordingly, the selective GLP-1 receptor antagonist Exendin-3 (300 nM) caused a significant reduction (p < 0.01) of the GLP-1 maximum effect only in GK rats [GLP-1 + Exendin-3 (W) = 107.79% ± 13.48, n = 8–7 strips/4 rats and GLP-1 + Exendin-3 (GK) = 55.08% ± 9.25, n = 5–6 strips/4 rats], without significant effect in potency (Figure 5). To rule out the involvement of acetylcholine, the primary neurotransmitter responsible for gastrointestinal contractility, we also conducted CR experiments for Liraglutide, in the presence and absence of 10 µM atropine, a non-selective muscarinic receptor antagonist, obviously using non-pre-contracted gastric fundus strips isolated from Wistar rats. Our findings suggest that the Liraglutide-induced tonic contractions are not of cholinergic origin (see Figure S2a,b).

4. Discussion

In the present study, we have focused on the pharmacological mechanism underlying the modulation of gastric fundus motility by GLP-1RA, since peripheral GLP-1 is known to delay gastric emptying and small intestinal motility and secretion through an interaction with brain centers or afferent neural pathways relaying to the vagal motor nuclei [29,30,31]. We have also evaluated the alterations of gastric fundus motility induced by this class of incretin mimetics in a rat model of T2DM, the GK rat.
In rats treated with Liraglutide (200 μg/kg, twice daily for fourteen days), we demonstrated that administration of this long-acting GLP-1A improved the lipidic profile in diabetic rats and caused a significant reduction in caloric intake and body weight in both diabetic and non-diabetic Liraglutide-treated animals. Our results are similar to those previously reported with obese and/or diabetic rats treated with Liraglutide [24,25,32]. Unexpectedly, Liraglutide reduced the relaxation induced by SNP and NA in gastric fundus strips isolated from treated animals, compared to non-treated groups, with effects differing depending on the presence or absence of diabetes. With respect to NA-induced relaxation, Liraglutide treatment significantly reduced both the intrinsic activity and potency of NA in gastric fundus strips from control and diabetic rats. Regarding NO-induced relaxation, no significant changes were observed in the maximum relaxation to SNP; only a significant reduction in its potency was observed, caused by the disease and treatment. These pharmacological findings should indicate a greater increase in NO concentration to achieve the same level of efficacy. However, no differences were observed in the levels of total and phosphorylated forms of nNOS in either animal model. This contrasts with our histological findings in the rat gastric fundus, which revealed GLP-1R-positive staining in ganglia, suggesting a neuronal role for GLP-1R, and with previous reports indicating the loss of nitrergic neurons—and consequently reduced NO synthesis—in the gastrointestinal tract of various diabetic models [33,34,35], as well as studies showing NO-dependent inhibition of gastrointestinal motility by GLP-1 [18,19,20,21,22]. Such differences should be further investigated.
In view of these results, which question the role of GLP-1RA as facilitators of NO and sympathetic-induced gastric emptying, we decided to verify whether GLP-1RA would induce contraction instead, under the same experimental conditions. Interestingly, the tested GLP-1RA caused tonic concentration-dependent contraction of the gastric fundus isolated from Wistar and GK rats after a CCh-induced pre-contraction, Liraglutide being the most potent of the agonists tested, but the least effective, especially compared to the short-acting GLP-1RA exenatide.
Additionally, native GLP-1 showed more intrinsic activity in diabetic rats than in control ones, which is consistent with the observed trend for higher GLP-1R protein levels in the gastric fundus of GK rats. This may reflect a compensatory mechanism resulting from lower endogenous GLP-1 production in this animal model, as already reported [36]. However, other reports point to a decrease in disease states. For instance, reduced GLP-1R expression in gastric glands of patients with T2DM was observed by Broide et al. [37] and significantly lower Glp-1r expression levels in endothelial and smooth muscle cells were observed in obese type 2 diabetic db/db mice compared to non-diabetic db/m mice [38]. Very little data has been reported regarding the effects of GLP-1RA treatment on receptor expression that can support our reduction in GLP-1R protein levels by Liraglutide treatment, regardless of the presence or absence of diabetes. In a report by Sanada et al. [39] with the long-acting incretin mimetic dulaglutide, Glp-1r expression varies depending on whether it is an early or late pharmacological intervention. Indeed, Glp-1r expression in the abdominal aorta of streptozotocin-induced diabetic Apoe knockout mice was reduced, becoming comparable to non-diabetic mice if an early intervention dulaglutide (twice weekly for 8 weeks) treatment was performed. However, after a late treatment (twice weekly for 16 weeks), Glp-1r expression was lower in controls, and dulaglutide failed to recover it. On the other hand, the reduced GLP-1R protein levels observed in Liraglutide-treated Wistar and GK rats may help explain the tachyphylaxis described for long-acting GLP-1RA on gastric emptying [11,12]. Although GLP-1RAs initially inhibit gastric motility, their efficacy in maintaining this effect diminishes over time, and the underlying mechanisms responsible for this attenuation remain unclear. All GLP-1RAs effectively slowed gastric emptying in the early stages of treatment, but within 1–2 weeks, their efficacy waned, and it has been suggested to be associated with a decline of Glp-1r mRNA expression in neurons of the nucleus of the solitary tract which plays a fundamental role in regulating metabolism and gastric motility [40].
Finally, the contractile effects of GLP-1RA on the rat gastric fundus are not mediated by acetylcholine—the primary excitatory neurotransmitter in the gastrointestinal tract—as they are unaffected by the non-selective muscarinic receptor antagonist atropine and persist in the presence of pre-contraction induced by CCh. On the other hand, they are unexpected, considering the GLP-1RA vasorelaxation via endothelium-derived NO, which has been described in the cardiovascular system. Despite the ongoing debate regarding the presence of GLP-1R in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), several studies have demonstrated that direct exposure of VSMCs and ECs to native GLP-1 or GLP-1RA increases eNOS phosphorylation and NO production via a 5′AMP-activated protein kinase (AMPK)-dependent mechanism [17,41]. However, and according to other studies exploring the location of GLP-1R in human, rat and monkey [23,37,42], our histological analysis of the rat gastric fundus did not reveal GLP-1R immunoreactivity in smooth muscle cells nor endothelial cells of arterioles, thus providing no anatomical evidence to support such a relaxation effect. In fact, it has been recognized that GLP-1 modulates antro-pyloro-duodenal motility by stimulating pyloric motility and inhibiting antral motility [43]. The precise mechanism by which GLP-1 does this is not entirely clear.
Moreover, our data indicates that the contractile response of the gastric fundus to GLP-1RA involves both GLP-1R-dependent and -independent mechanisms, as evidenced by the significant reduction in the maximum effect of GLP-1 following incubation with the selective GLP-1R antagonist Exendin-3 in GK rats, but not in control animals. This would not be unprecedented since some GLP-1 effects in the cardiovascular system were suggested to be independent of this receptor [5]. Supporting this, animal studies have shown no direct effect of GLP-1 on gastric smooth muscle cells [44]. This reinforces the notion that suppression of vagal input from neural afferents is involved in the inhibition of gastric motor functions by exogenous GLP-1 [29,30,31]. Additional studies are needed to elucidate the mechanisms mediating GLP-1-induced gastric motor effects in vivo.
This study provides evidence of a contractile response to GLP-1RA in rat gastric fundus that is partially mediated by GLP-1R. In diabetic rats, the intrinsic activity of GLP-1 was higher, which may be attributed to a compensatory mechanism induced by T2DM, as suggested by a trend toward increased levels of receptor proteins. The significant reduction in GLP-1R protein levels following Liraglutide treatment was associated with diminished NA- and SNP-mediated relaxations, contributing to an overall increase in gastric fundus tone. Thus, given the satiating effect of gastric distension [45], our results suggest that the increased gastric fundus tone induced by this long-acting GLP-1RA may reduce gastric accommodation during a meal. These mechanisms may contribute to the GLP-1RA-induced reduction in food intake, alongside the well-established central pathways, and support the therapeutic benefits of GLP-1RAs in the management of obesity and T2DM.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/diabetology6090096/s1, Figure S1: Representative GLP-1R immunohistochemical findings on isolated gastric fundus from Wistar control rats (WC), GK control rats (GKC) and GK rats treated with Liraglutide (GKL); Figure S2a,b: Representative recordings of cumulative CR curve for Liraglutide in gastric fundus strips isolated from Wistar rats, in the presence and absence of atropine.

Author Contributions

Conducted functional studies: A.C.C., B.M. and S.S. Conducted in vivo experiments: A.C.C., B.M. and P.M. Conducted biochemical and tissue analysis: A.C.C. and B.M. Performed data analysis: A.C.C. and B.M. Contributed to the writing of the manuscript: A.C.C., B.M., P.M. and S.S. Data discussion and the final revision of the manuscript: R.S., C.F.-R., P.M. and S.S. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the Portuguese Foundation for Science and Technology (PEst UIDB/04539/Base/2020 and UIDP/04539/Programatico/2020) and by the Faculty of Medicine, University of Coimbra.

Institutional Review Board Statement

The animal study protocol was approved by the Institutional Review Board of Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, (protocol code 13-2018 and date of approval 22 March 2019) for studies involving animals.

Informed Consent Statement

Not applicable.

Data Availability Statement

The quantitative data presented in this study are available on reasonable request from the corresponding author. The raw data supporting the conclusions of this article, obtained in isolated vessels and histology are available as supplementary data.

Acknowledgments

The authors gratefully acknowledge the scientific advice of Lina Carvalho and the technical support in the immunohistochemistry studies of Lia Neves from the Institute of Pathological Anatomy, Faculty of Medicine, University of Coimbra, Portugal.

Conflicts of Interest

The authors have no disclosures to make.

Abbreviations

AUC—Area under the curve; BCA—Bicinchoninic acid; CCh—Carbachol; CR—concentration-response; DPP-4—dipeptidyl peptidase 4; DPX—mixture of distyrene, a plasticiser and xylene; ECL—enhanced chemiluminescence; Emax—maximum contractile/relaxation response; eNOS—Endothelial nitric oxide synthase; FELASA—Federation of Laboratory Animal Science Associations; GK—Goto-Kakisaki; GKC—non-treated Goto-Kakisaki rats; GKL—Goto-Kakisaki rats treated with Liraglutide; GLP-1—Glucagon-like peptide 1; GLP-1R—Glucagon-like peptide 1 receptor; GLP-1RA—Glucagon-like peptide 1 receptor agonist; IDF—International Diabetes Federation; ipITT—intraperitoneal Insulin tolerance test; NA—Noradrenaline; nNOS—Neuronal nitric oxide synthase; NO—Nitric oxide; pEC50—negative logarithm of the molar concentration of agonist inducing half maximum response; PI3K—Phosphoinositide 3-kinase; PKB/Akt—Protein Kinase B; p-nNOS—Neuronal nitric oxide synthase phosphorylated; PVDF—Polyvinylidene fluoride; s.c.—subcutaneous; SDS-PAGE—sodium dodecyl sulfate–polyacrylamide gel electrophoresis; SNP—Sodium Nitroprusside; TBS-T: Tris-buffered saline with Tween 20; T2DM—Type 2 Diabetes Mellitus; W—Wistar; WC—non-treated Wistar rats; WL—Wistar rats treated with Liraglutide.

References

  1. International Diabetes Federation. IDF Diabetes Atlas, 11th ed.; International Diabetes Federation: Brussels, Belgium, 2025. [Google Scholar]
  2. ElSayed, N.A.; Aleppo, G.; Aroda, V.R.; Bannuru, R.R.; Brown, F.M.; Bruemmer, D.; Collins, B.S.; Hilliard, M.E.; Isaacs, D.; Johnson, E.L.; et al. 3. Prevention or Delay of Type 2 Diabetes and Associated Comorbidities: Standards of Care in Diabetes-2023. Diabetes Care 2023, 46 (Suppl. S1), S41–S48. [Google Scholar] [CrossRef] [PubMed]
  3. Tasyurek, H.M.; Altunbas, H.A.; Balci, M.K.; Sanlioglu, S. Incretins: Their physiology and application in the treatment of diabetes mellitus. Diabetes Metab. Res. Rev. 2014, 30, 354–371. [Google Scholar] [CrossRef] [PubMed]
  4. Li, J.; Zheng, J.; Wang, S.; Lau, H.K.; Fathi, A.; Wang, Q. Cardiovascular Benefits of Native GLP-1 and its Metabolites: An Indicator for GLP-1-Therapy Strategies. Front. Physiol. 2017, 8, 15. [Google Scholar] [CrossRef] [PubMed]
  5. Ban, K.; Noyan-Ashraf, M.H.; Hoefer, J.; Bolz, S.S.; Drucker, D.J.; Husain, M. Cardioprotective and vasodilatory actions of glucagon-like peptide 1 receptor are mediated through both glucagon-like peptide 1 receptor-dependent and -independent pathways. Circulation 2008, 117, 2340–2350. [Google Scholar] [CrossRef]
  6. Nauck, M.A.; Meier, J.J. MANAGEMENT OF ENDOCRINE DISEASE: Are all GLP-1 agonists equal in the treatment of type 2 diabetes? Eur. J. Endocrinol. 2019, 181, R211–R234. [Google Scholar] [CrossRef]
  7. Jacobsen, L.V.; Flint, A.; Olsen, A.K.; Ingwersen, S.H. Liraglutide in Type 2 Diabetes Mellitus: Clinical Pharmacokinetics and Pharmacodynamics. Clin. Pharmacokinet. 2016, 55, 657–672. [Google Scholar] [CrossRef]
  8. Knudsen, L.B.; Lau, J. The Discovery and Development of Liraglutide and Semaglutide. Front. Endocrinol. 2019, 10, 155. [Google Scholar] [CrossRef]
  9. Secher, A.; Jelsing, J.; Baquero, A.F.; Hecksher-Sorensen, J.; Cowley, M.A.; Dalboge, L.S.; Hansen, G.; Grove, K.L.; Pyke, C.; Raun, K.; et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J. Clin. Investig. 2014, 124, 4473–4488. [Google Scholar] [CrossRef]
  10. van Bloemendaal, L.; Veltman, D.J.; Ten Kulve, J.S.; Groot, P.F.; Ruhe, H.G.; Barkhof, F.; Sloan, J.H.; Diamant, M.; Ijzerman, R.G. Brain reward-system activation in response to anticipation and consumption of palatable food is altered by glucagon-like peptide-1 receptor activation in humans. Diabetes Obes. Metab. 2015, 17, 878–886. [Google Scholar] [CrossRef]
  11. Ard, J.; Fitch, A.; Fruh, S.; Herman, L. Weight Loss and Maintenance Related to the Mechanism of Action of Glucagon-Like Peptide 1 Receptor Agonists. Adv. Ther. 2021, 38, 2821–2839. [Google Scholar] [CrossRef]
  12. van Can, J.; Sloth, B.; Jensen, C.B.; Flint, A.; Blaak, E.E.; Saris, W.H. Effects of the once-daily GLP-1 analog liraglutide on gastric emptying, glycemic parameters, appetite and energy metabolism in obese, non-diabetic adults. Int. J. Obes. 2014, 38, 784–793. [Google Scholar] [CrossRef]
  13. Nielsen, L.L.; Young, A.A.; Parkes, D.G. Pharmacology of exenatide (synthetic exendin-4): A potential therapeutic for improved glycemic control of type 2 diabetes. Regul. Pept. 2004, 117, 77–88. [Google Scholar] [CrossRef]
  14. Drucker, D.J. The biology of incretin hormones. Cell Metab. 2006, 3, 153–165. [Google Scholar] [CrossRef]
  15. Andersen, A.; Lund, A.; Knop, F.K.; Vilsboll, T. Glucagon-like peptide 1 in health and disease. Nat. Rev. Endocrinol. 2018, 14, 390–403. [Google Scholar] [CrossRef]
  16. Campbell, J.E.; Drucker, D.J. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 2013, 17, 819–837. [Google Scholar] [CrossRef] [PubMed]
  17. Zhong, J.; Rao, X.; Rajagopalan, S. An emerging role of dipeptidyl peptidase 4 (DPP4) beyond glucose control: Potential implications in cardiovascular disease. Atherosclerosis 2013, 226, 305–314. [Google Scholar] [CrossRef] [PubMed]
  18. Amato, A.; Cinci, L.; Rotondo, A.; Serio, R.; Faussone-Pellegrini, M.S.; Vannucchi, M.G.; Mule, F. Peripheral motor action of glucagon-like peptide-1 through enteric neuronal receptors. Neurogastroenterol. Motil. 2010, 22, 664–e203. [Google Scholar] [CrossRef]
  19. Amato, A.; Baldassano, S.; Liotta, R.; Serio, R.; Mule, F. Exogenous glucagon-like peptide 1 reduces contractions in human colon circular muscle. J. Endocrinol. 2014, 221, 29–37. [Google Scholar] [CrossRef]
  20. Marathe, C.S.; Rayner, C.K.; Jones, K.L.; Horowitz, M. Effects of GLP-1 and incretin-based therapies on gastrointestinal motor function. Exp. Diabetes Res. 2011, 2011, 279530. [Google Scholar] [CrossRef]
  21. Rotondo, A.; Amato, A.; Lentini, L.; Baldassano, S.; Mule, F. Glucagon-like peptide-1 relaxes gastric antrum through nitric oxide in mice. Peptides 2011, 32, 60–64. [Google Scholar] [CrossRef]
  22. Schirra, J.; Nicolaus, M.; Woerle, H.J.; Struckmeier, C.; Katschinski, M.; Goke, B. GLP-1 regulates gastroduodenal motility involving cholinergic pathways. Neurogastroenterol. Motil. 2009, 21, 609-e22. [Google Scholar] [CrossRef] [PubMed]
  23. Pyke, C.; Heller, R.S.; Kirk, R.K.; Orskov, C.; Reedtz-Runge, S.; Kaastrup, P.; Hvelplund, A.; Bardram, L.; Calatayud, D.; Knudsen, L.B. GLP-1 receptor localization in monkey and human tissue: Novel distribution revealed with extensively validated monoclonal antibody. Endocrinology 2014, 155, 1280–1290. [Google Scholar] [CrossRef] [PubMed]
  24. Sturis, J.; Gotfredsen, C.F.; Romer, J.; Rolin, B.; Ribel, U.; Brand, C.L.; Wilken, M.; Wassermann, K.; Deacon, C.F.; Carr, R.D.; et al. GLP-1 derivative liraglutide in rats with beta-cell deficiencies: Influence of metabolic state on beta-cell mass dynamics. Br. J. Pharmacol. 2003, 140, 123–132. [Google Scholar] [CrossRef] [PubMed]
  25. Nagakubo, D.; Shirai, M.; Nakamura, Y.; Kaji, N.; Arisato, C.; Watanabe, S.; Takasugi, A.; Asai, F. Prophylactic effects of the glucagon-like Peptide-1 analog liraglutide on hyperglycemia in a rat model of type 2 diabetes mellitus associated with chronic pancreatitis and obesity. Comp. Med. 2014, 64, 121–127. [Google Scholar]
  26. Riazi-Farzad, B.; Nicholls, P.J.; Sewell, R.D. Sensitivity differences to 5-HT and carbachol in subsections of the isolated rat stomach fundus strip: An improved preparation. J. Pharmacol. Toxicol. Methods 1996, 35, 217–221. [Google Scholar] [CrossRef]
  27. Vane, J.R. A sensitive method for the assay of 5-hydroxytryptamine. Br. J. Pharmacol. 1997, 120 (Suppl. S4), 142–147; discussion 140–141. [Google Scholar] [CrossRef]
  28. Rocha, B.S.; Gago, B.; Barbosa, R.M.; Cavaleiro, C.; Laranjinha, J. Ethyl nitrite is produced in the human stomach from dietary nitrate and ethanol, releasing nitric oxide at physiological pH: Potential impact on gastric motility. Free Radic. Biol. Med. 2015, 82, 160–166. [Google Scholar] [CrossRef]
  29. Tolessa, T.; Gutniak, M.; Holst, J.J.; Efendic, S.; Hellstrom, P.M. Glucagon-like peptide-1 retards gastric emptying and small bowel transit in the rat: Effect mediated through central or enteric nervous mechanisms. Dig. Dis. Sci. 1998, 43, 2284–2290. [Google Scholar] [CrossRef]
  30. Wettergren, A.; Wojdemann, M.; Holst, J.J. Glucagon-like peptide-1 inhibits gastropancreatic function by inhibiting central parasympathetic outflow. Am. J. Physiol. 1998, 275, G984–G992. [Google Scholar] [CrossRef]
  31. Imeryuz, N.; Yegen, B.C.; Bozkurt, A.; Coskun, T.; Villanueva-Penacarrillo, M.L.; Ulusoy, N.B. Glucagon-like peptide-1 inhibits gastric emptying via vagal afferent-mediated central mechanisms. Am. J. Physiol. 1997, 273, G920–G927. [Google Scholar] [CrossRef]
  32. Larsen, P.J.; Fledelius, C.; Knudsen, L.B.; Tang-Christensen, M. Systemic administration of the long-acting GLP-1 derivative NN2211 induces lasting and reversible weight loss in both normal and obese rats. Diabetes 2001, 50, 2530–2539. [Google Scholar] [CrossRef]
  33. Bagyanszki, M.; Bodi, N. Diabetes-related alterations in the enteric nervous system and its microenvironment. World J. Diabetes 2012, 3, 80–93. [Google Scholar] [CrossRef]
  34. Cellek, S. Point of NO return for nitrergic nerves in diabetes: A new insight into diabetic complications. Curr. Pharm. Des. 2004, 10, 3683–3695. [Google Scholar] [CrossRef] [PubMed]
  35. Lu, H.L.; Huang, X.; Wu, Y.S.; Zhang, C.M.; Meng, X.M.; Liu, D.H.; Kim, Y.C.; Xu, W.X. Gastric nNOS reduction accompanied by natriuretic peptides signaling pathway upregulation in diabetic mice. World J. Gastroenterol. 2014, 20, 4626–4635. [Google Scholar] [CrossRef] [PubMed]
  36. Eickhoff, H.; Louro, T.M.; Matafome, P.N.; Vasconcelos, F.; Seica, R.M.; Castro, E.S.F. Amelioration of glycemic control by sleeve gastrectomy and gastric bypass in a lean animal model of type 2 diabetes: Restoration of gut hormone profile. Obes. Surg. 2015, 25, 7–18. [Google Scholar] [CrossRef]
  37. Broide, E.; Bloch, O.; Ben-Yehudah, G.; Cantrell, D.; Shirin, H.; Rapoport, M.J. Reduced GLP-1R expression in gastric glands of patients with type 2 diabetes mellitus. J. Clin. Endocrinol. Metab. 2014, 99, E1691–E1695. [Google Scholar] [CrossRef]
  38. Kimura, T.; Obata, A.; Shimoda, M.; Okauchi, S.; Hirukawa, H.; Kohara, K.; Kinoshita, T.; Nogami, Y.; Nakanishi, S.; Mune, T.; et al. Decreased glucagon-like peptide 1 receptor expression in endothelial and smooth muscle cells in diabetic db/db mice: TCF7L2 is a possible regulator of the vascular glucagon-like peptide 1 receptor. Diabetes Vasc. Dis. Res. 2017, 14, 540–548. [Google Scholar] [CrossRef]
  39. Sanada, J.; Obata, A.; Obata, Y.; Fushimi, Y.; Shimoda, M.; Kohara, K.; Nakanishi, S.; Mune, T.; Kaku, K.; Kaneto, H. Dulaglutide exerts beneficial anti atherosclerotic effects in ApoE knockout mice with diabetes: The earlier, the better. Sci. Rep. 2021, 11, 1425. [Google Scholar] [CrossRef]
  40. Wang, P.; Mamillapalli, R.; Zhu, W.; Chan, O. Long-Term Use of GLP-1 Receptor Agonists Alter GLP-1 Receptor mRNA Expression in Hindbrain Pathways That Regulate Gastric Motility in Mice. Diabetes 2018, 67 (Suppl. S1), 1088–P. [Google Scholar] [CrossRef]
  41. Almutairi, M.B.; Al Batran, R.; Ussher, J.R. Glucagon-like peptide-1 receptor action in the vasculature. Peptides 2019, 111, 26–32. [Google Scholar] [CrossRef]
  42. Bloch, O.; Broide, E.; Ben-Yehudah, G.; Cantrell, D.; Shirin, H.; Rapoport, M.J. Nutrient induced type 2 and chemical induced type 1 experimental diabetes differently modulate gastric GLP-1 receptor expression. J. Diabetes Res. 2015, 2015, 561353. [Google Scholar] [CrossRef] [PubMed]
  43. Schirra, J.; Nicolaus, M.; Roggel, R.; Katschinski, M.; Storr, M.; Woerle, H.J.; Goke, B. Endogenous glucagon-like peptide 1 controls endocrine pancreatic secretion and antro-pyloro-duodenal motility in humans. Gut 2006, 55, 243–251. [Google Scholar] [CrossRef] [PubMed]
  44. Rodier, G.; Magous, R.; Mochizuki, T.; Martinez, J.; Nguyen, D.L.; Bali, J.P.; Bataille, D.; Jarrousse, C. Effect of glicentin, oxyntomodulin and related peptides on isolated gastric smooth muscle cells. Pflug. Arch. 1997, 434, 729–734. [Google Scholar] [CrossRef]
  45. Janssen, P.; Vanden Berghe, P.; Verschueren, S.; Lehmann, A.; Depoortere, I.; Tack, J. Review article: The role of gastric motility in the control of food intake. Aliment. Pharmacol. Ther. 2011, 33, 880–894. [Google Scholar] [CrossRef]
Figure 1. Representative recordings of isometric cumulative CR curves. Representative recording of isometric cumulative CR curve for GLP-1 in gastric fundus strips isolated from non-treated Wistar rats, pre-contracted with 5µM of carbachol (CCh).
Figure 1. Representative recordings of isometric cumulative CR curves. Representative recording of isometric cumulative CR curve for GLP-1 in gastric fundus strips isolated from non-treated Wistar rats, pre-contracted with 5µM of carbachol (CCh).
Diabetology 06 00096 g001
Figure 2. Evaluation of Liraglutide effects on biological and biochemical parameters during the treatment period, namely body weight gain (A), caloric intake (B), fasting glycemia (C), area under the curve (AUC) of intraperitoneal insulin tolerance test (ITT) (D), triglycerides (E) and total cholesterol (F). WC = Wistar rat injected with saline; WL = Wistar rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days); GKC = Goto-Kakizaki rat injected with saline; GKL = Goto-Kakizaki rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days). Results are presented as mean ± S.E.M.; vertical bars indicate the S.E.M.; the number of animals per group ranged from 4 to 7 (n = 4–7/group); statistical differences among experimental groups were assessed using the Kruskal–Wallis test. Comparisons between initial and final values of biochemical parameters within the same animal were performed using the Wilcoxon signed-rank test for paired data. * p < 0.05, ** p < 0.01 vs. WC; # p < 0.05, ## p < 0.01 vs. GKC; § p < 0.05 vs. Initial.
Figure 2. Evaluation of Liraglutide effects on biological and biochemical parameters during the treatment period, namely body weight gain (A), caloric intake (B), fasting glycemia (C), area under the curve (AUC) of intraperitoneal insulin tolerance test (ITT) (D), triglycerides (E) and total cholesterol (F). WC = Wistar rat injected with saline; WL = Wistar rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days); GKC = Goto-Kakizaki rat injected with saline; GKL = Goto-Kakizaki rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days). Results are presented as mean ± S.E.M.; vertical bars indicate the S.E.M.; the number of animals per group ranged from 4 to 7 (n = 4–7/group); statistical differences among experimental groups were assessed using the Kruskal–Wallis test. Comparisons between initial and final values of biochemical parameters within the same animal were performed using the Wilcoxon signed-rank test for paired data. * p < 0.05, ** p < 0.01 vs. WC; # p < 0.05, ## p < 0.01 vs. GKC; § p < 0.05 vs. Initial.
Diabetology 06 00096 g002
Figure 3. Reduced gastric fundus GLP-1R protein levels in Wistar and GK rats after Liraglutide treatment calculated by Western blotting as a percentage of the WC group/Calnexin. A representative Western blot image is shown in (A), and representative GLP-1R immunohistochemical findings in the rat gastric fundus are presented in (B). (A) WC = Wistar rat injected with saline; WL = Wistar rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days); GKC = Goto-Kakizaki rat injected with saline; GKL = Goto-Kakizaki rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days). Results are presented as mean ± S.E.M.; vertical bars indicate the S.E.M.; n = 4–7 animals/group; statistical differences were evaluated by the Kruskal–Wallis test. * p < 0.05 vs. WC; # p < 0.05, ## p < 0.01 vs. GKC. (B) Representative HE (panel 1) and GLP-1R immunohistochemical findings (panels 24) on isolated gastric fundus from Wistar rats treated with Liraglutide. GLP-1R immunostaining appears in brown, counterstained with hematoxylin. GLP-1R positive cells in ganglion cells (a) and in endothelial cells of capillaries (b) but not on smooth muscle cells of muscularis mucosa (c) and endothelial cells of arterioles (d). Scale bar: 100 µm in panels 13 and 50 µm in panel 4.
Figure 3. Reduced gastric fundus GLP-1R protein levels in Wistar and GK rats after Liraglutide treatment calculated by Western blotting as a percentage of the WC group/Calnexin. A representative Western blot image is shown in (A), and representative GLP-1R immunohistochemical findings in the rat gastric fundus are presented in (B). (A) WC = Wistar rat injected with saline; WL = Wistar rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days); GKC = Goto-Kakizaki rat injected with saline; GKL = Goto-Kakizaki rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days). Results are presented as mean ± S.E.M.; vertical bars indicate the S.E.M.; n = 4–7 animals/group; statistical differences were evaluated by the Kruskal–Wallis test. * p < 0.05 vs. WC; # p < 0.05, ## p < 0.01 vs. GKC. (B) Representative HE (panel 1) and GLP-1R immunohistochemical findings (panels 24) on isolated gastric fundus from Wistar rats treated with Liraglutide. GLP-1R immunostaining appears in brown, counterstained with hematoxylin. GLP-1R positive cells in ganglion cells (a) and in endothelial cells of capillaries (b) but not on smooth muscle cells of muscularis mucosa (c) and endothelial cells of arterioles (d). Scale bar: 100 µm in panels 13 and 50 µm in panel 4.
Diabetology 06 00096 g003
Figure 4. Effects of liraglutide treatment on gastric fundus smooth muscle relaxation induced by (A) sodium nitroprusside (SNP) and (B) noradrenaline (NA), as well as on (C) nNOS and p-nNOS protein levels of the same Wistar and GK rats, quantified by Western blotting and expressed as a percentage relative to the WC group normalized to Calnexin. Representative Western blot images are shown. WC = Wistar rat injected with saline; WL = Wistar rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days); GKC = Goto-Kakizaki rat injected with saline; GKL = Goto-Kakizaki rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days). Results are presented as mean ± S.E.M.; vertical bars indicate the S.E.M.; n = 4–7 animals/group; statistical differences were evaluated by the Kruskal–Wallis test (for significant differences in the functional study see Table 1).
Figure 4. Effects of liraglutide treatment on gastric fundus smooth muscle relaxation induced by (A) sodium nitroprusside (SNP) and (B) noradrenaline (NA), as well as on (C) nNOS and p-nNOS protein levels of the same Wistar and GK rats, quantified by Western blotting and expressed as a percentage relative to the WC group normalized to Calnexin. Representative Western blot images are shown. WC = Wistar rat injected with saline; WL = Wistar rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days); GKC = Goto-Kakizaki rat injected with saline; GKL = Goto-Kakizaki rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days). Results are presented as mean ± S.E.M.; vertical bars indicate the S.E.M.; n = 4–7 animals/group; statistical differences were evaluated by the Kruskal–Wallis test (for significant differences in the functional study see Table 1).
Diabetology 06 00096 g004
Figure 5. Contractile responses of Wistar (A) and GK (B) rat gastric fundus strips to GLP-1 in the absence and in the presence of 300 nM Exendin-3. Results are presented as mean ± S.E.M; vertical bars indicate the S.E.M; n = 8–5 strips from 4 rats/group. Statistical differences were evaluated by Mann–Whitney test. * p < 0.05, ** p < 0.01 vs. GLP-1.
Figure 5. Contractile responses of Wistar (A) and GK (B) rat gastric fundus strips to GLP-1 in the absence and in the presence of 300 nM Exendin-3. Results are presented as mean ± S.E.M; vertical bars indicate the S.E.M; n = 8–5 strips from 4 rats/group. Statistical differences were evaluated by Mann–Whitney test. * p < 0.05, ** p < 0.01 vs. GLP-1.
Diabetology 06 00096 g005
Table 1. Emax and pEC50 values for SNP and NA in gastric fundus isolated from Wistar and GK rats treated and non-treated with Liraglutide.
Table 1. Emax and pEC50 values for SNP and NA in gastric fundus isolated from Wistar and GK rats treated and non-treated with Liraglutide.
Sodium Nitroprusside (SNP)Noradrenaline (NA)
Emax (%CCh)pEC50s/nEmax (%CCh)pEC50s/n
WC64.64 ± 7.066.26 ± 0.1614/485.38 ± 5.216.12 ± 0.0916/4
WL43.60 ± 4.926.12 ± 0.1215/461.63 ± 5.22 *5.7 ± 0.15 *16/4
GKC55.75 ± 8.615.85 ± 0.1015/460.44 ± 4.86 *5.62 ± 0.06 **14/4
GKL47.42 ± 5.125.22 ± 0.09 **** ###16/472.20 ± 6.105.27 ± 0.07 ***16/4
Emax = maximum relaxation in percentage of inhibition of CCh induced contraction; pEC50 = negative logarithm of the molar concentration of SNP/NA inducing half maximum relaxation; s/n = number of gastric fundus strips tested (s)/number of rats (n) from whom tissues were obtained; WC = Wistar rat injected with saline; WL = Wistar rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days); GKC = Goto-Kakizaki rat injected with saline; GKL = Goto-Kakizaki rat injected with Liraglutide (200 μg/kg s.c., twice daily for 14 days). Results are presented as mean ± S.E.M of the indicated tissue strips isolated from n animals. Statistical differences were evaluated by the Kruskal–Wallis test. * p < 0.05, ** p < 0.01, *** p < 0.005, **** p < 0.001 vs. WC; ### p < 0.005 vs. GKC.
Table 2. Emax and pEC50 values for GLP-1RA in the gastric fundus isolated from Wistar and GK rats.
Table 2. Emax and pEC50 values for GLP-1RA in the gastric fundus isolated from Wistar and GK rats.
Emax (mN)pEC50s/n
WistarGKWistarGKWistarGK
GLP-15.61 ± 0.699.09 ± 1.44 *8.33 ± 0.168.58 ± 0.1616/412/4
Exenatide11.67 ± 1.08 ####10.37 ± 1.998.78 ± 0.168.92 ± 0.2111/312/3
Liraglutide4.69 ± 0.81 §§§§3.76 ± 1.01 # §§9.62 ± 0.24 #### §9.06 ± 0.5113/45/3
Emax = maximum contraction in mN of tension; pEC50 = negative logarithm of the molar concentration of agonist inducing half maximum contraction; s/n = number of gastric fundus strips tested (s)/number of rats (n) from whom tissues were obtained. Results are presented as mean ± S.E.M. Statistical differences were evaluated by the Kruskal–Wallis test and Mann–Whitney test. * p < 0.05 vs. Wistar; # p < 0.05, #### p < 0.001 vs. GLP-1; § p < 0.05, §§ p < 0.01, §§§§ p < 0.001 vs. Exenatide.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Carrêlo, A.C.; Martins, B.; Seiça, R.; Fontes-Ribeiro, C.; Matafome, P.; Silva, S. Liraglutide Increases Gastric Fundus Tonus and Reduces Food Intake in Type 2 Diabetic Rats. Diabetology 2025, 6, 96. https://doi.org/10.3390/diabetology6090096

AMA Style

Carrêlo AC, Martins B, Seiça R, Fontes-Ribeiro C, Matafome P, Silva S. Liraglutide Increases Gastric Fundus Tonus and Reduces Food Intake in Type 2 Diabetic Rats. Diabetology. 2025; 6(9):96. https://doi.org/10.3390/diabetology6090096

Chicago/Turabian Style

Carrêlo, Ana Catarina, Beatriz Martins, Raquel Seiça, Carlos Fontes-Ribeiro, Paulo Matafome, and Sónia Silva. 2025. "Liraglutide Increases Gastric Fundus Tonus and Reduces Food Intake in Type 2 Diabetic Rats" Diabetology 6, no. 9: 96. https://doi.org/10.3390/diabetology6090096

APA Style

Carrêlo, A. C., Martins, B., Seiça, R., Fontes-Ribeiro, C., Matafome, P., & Silva, S. (2025). Liraglutide Increases Gastric Fundus Tonus and Reduces Food Intake in Type 2 Diabetic Rats. Diabetology, 6(9), 96. https://doi.org/10.3390/diabetology6090096

Article Metrics

Back to TopTop