Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Articles

Page Path
HOME > Diabetes Metab J > Volume 49(3); 2025 > Article
Review
Basic and Translational Research Glucagon-Like Peptide-1 and Hypothalamic Regulation of Satiation: Cognitive and Neural Insights from Human and Animal Studies
Joon Seok Park1orcid, Kyu Sik Kim2, Hyung Jin Choi1,2,3,4,5,6orcidcorresp_icon
Diabetes & Metabolism Journal 2025;49(3):333-347.
DOI: https://doi.org/10.4093/dmj.2025.0106
Published online: May 1, 2025
  • 585 Views
  • 70 Download

1Department of Medicine, Seoul National University College of Medicine, Seoul, Korea

2Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea

3Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea

4Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea

5Wide River Institute of Immunology, Seoul National University, Hongcheon, Korea

6Department of Brain and Cognitive Sciences, Seoul National University, Seoul, Korea

corresp_icon Corresponding author: Hyung Jin Choi orcid Department of Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea E-mail: hjchoi@snu.ac.kr
• Received: February 9, 2025   • Accepted: April 16, 2025

Copyright © 2025 Korean Diabetes Association

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

next
  • Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have emerged as blockbuster drugs for treating metabolic diseases. Glucagon-like peptide-1 (GLP-1) plays a pivotal role in glucose homeostasis by enhancing insulin secretion, suppressing glucagon release, delaying gastric emptying, and acting on the central nervous system to regulate satiation and satiety. This review summarizes the discovery of GLP-1 and the development of GLP-1RAs, with a particular focus on their central mechanisms of action. Human neuroimaging studies demonstrate that GLP-1RAs influence brain activity during food cognition, supporting a role in pre-ingestive satiation. Animal studies on hypothalamic feed-forward regulation of hunger suggest that cognitive hypothalamic mechanisms may also contribute to satiation control. We highlight the brain mechanisms of GLP-1RA-induced satiation and satiety, including cognitive impacts, with an emphasis on animal studies of hypothalamic glucagon-like peptide-1 receptor (GLP-1R) and GLP-1R-expressing neurons. Actions in non-hypothalamic regions are also discussed. Additionally, we review emerging combination drugs and oral GLP-1RA formulations aimed at improving efficacy and patient adherence. In conclusion, the dorsomedial hypothalamus (DMH)—a key GLP-1RA target—mediates pre-ingestive cognitive satiation, while other hypothalamic GLP-1R neurons regulate diverse aspects of feeding behavior, offering potential therapeutic targets for obesity treatment.
• GLP-1RAs act centrally to induce satiation and satiety, and influence cognition.
• DMH GLP-1R neurons drive pre-ingestive, cognitive satiation via feed-forward control.
• Hypothalamic GLP-1R neurons in ARC, DMH, PVH, LH, and VMH shape distinct feeding traits.
• Hindbrain and septal GLP-1R neurons modulate aversion and satiety.
• Novel GLP-1RAs improve efficacy and show promise beyond obesity such as addiction.
Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have garnered significant attention in the global pharmaceutical market. Glucagon-like peptide-1 (GLP-1) is an incretin hormone derived from the transcription product of the proglucagon gene, primarily secreted by intestinal L-cells in response to nutrient intake [1]. It plays a critical role in glucose homeostasis, enhancing insulin secretion, suppressing glucagon release, and delaying gastric emptying.
In addition to its metabolic effects, GLP-1 acts centrally to maintain energy balance by activating glucagon-like peptide-1 receptors (GLP-1Rs) located in key regions of the brain. These include regions in the hypothalamus, brainstem, and mesolimbic reward pathways, which collectively regulate eating behavior such as food intake, satiation, and satiety. Additionally, the effects of GLP-1RAs on cognitive processes are currently being explored, broadening our understanding of their therapeutic mechanism in controlling metabolism.
This review will briefly cover the history of GLP-1 discovery and development of incretin-based drugs. Next, we will discuss human studies about the effects of GLP-1 on the central nervous system (CNS) and cognitive behavior, and animal studies that imply cognitive actions regulated by the hypothalamus. We will then go through the mechanism of GLP-1 acting on neural circuits within the CNS that regulate satiation and satiety, particularly focusing on regulation by the hypothalamus.
The discovery of secretin in 1902 was the hallmark for the initiation of gastrointestinal endocrinology [2]. In 1964, the development of insulin radioimmunoassay methods led to the discovery of the incretin effect, which demonstrated that oral glucose intake induces higher insulin secretion compared to intravenous (i.v.) glucose [3,4]. In 1973, Dupre et al. [5] proved that GIP, originally named ‘gastric inhibitory polypeptide,’ could explain this incretin effect by inducing glucose-stimulated insulin secretion, and renamed the peptide as ‘glucose-dependent insulinotropic polypeptide.’ Following the identification of GIP, the search for insulinotropic incretins persevered.
Meanwhile in 1982, Joel Habener’s group performed a cDNA study on anglerfish, a rich source of pancreatic islet hormone mRNAs. They revealed that the glucagon precursor gene encoded additional peptides beyond glucagon [6,7]. Subsequently, a study by Bell et al. [8] conducted cDNA analysis of preproglucagon in hamsters, which revealed that mammalian genes differ from those of anglerfish and identified two novel peptides, terming them glucagon-related peptides 1 and 2 (GLP-1, GLP-2).
GLP-1’s role as an insulinotropic incretin remained undetermined until GLP-1 (7-37), a truncated form of GLP-1, was identified by Mojsov et al. [9] as capable of stimulating insulin release in the rat pancreas, in contrast to GLP-1 (1-37), which did not alter insulin secretion. Holst et al. [10] further demonstrated that GLP-1 (7-37) extracted from pig intestine could stimulate insulin secretion in pig pancreas. In 1987, Kreymann et al. [11] performed studies in humans and confirmed GLP-1 (7-36) to be insulinotropic, suggesting its incretin nature. Indeed, i.v. infusion of GLP-1 (7-37) in subjects with and without diabetes elevated peak insulin levels by 10- and 3-fold respectively, and attenuated glucose levels [12]. These results suggested the possibility of GLP-1 as a treatment for diabetes, but limitations for therapeutic utility existed as endogenous GLP-1 had a short half-life in plasma of 1 to 2 minutes, degraded by the dipeptidyl-peptidase 4 (DPP-4) enzyme [13].
In 1992, Eng et al. [14] successfully isolated exendin-4 from the venom of the Gila monster (Heloderma suspectum), a lizard known for its infrequent but large meals weighing up to as much as one third of its body mass—suggesting the presence of adaptive metabolic mechanisms in the species [14,15]. Exendin-4, which shows 53% structural homology with GLP-1 (7-36), is as potent as endogenous GLP-1 in its insulinotropic effects via the mammalian GLP-1R. Strikingly, exendin-4 was stable against DPP-4 [16,17].
The isolation of exendin-4 led to the development of exenatide as the first U.S. Food and Drug Administration (FDA)-approved diabetes drug in 2005 by John Eng, with collaboration from Amylin Pharmaceutical (San Diego, CA, USA). Meanwhile, a group led by Knudsen at Novo Nordisk (Bagsværd, Denmark) employed the approach of fatty acid acylation to facilitate albumin binding and attenuate degradation [18]. The final product, liraglutide (Victoza, Novo Nordisk), proved its effects in type 2 diabetes mellitus (T2DM) and was approved by the FDA in 2010 [19]. Liraglutide (Saxenda, Novo Nordisk) was further developed with a higher dosage than Victoza for chronic weight management, and was approved by the FDA in 2014, proving its weight loss effects in numerous clinical studies performed worldwide [20,21].
The success of clinical trials with exenatide and liraglutide sparked growing interest in therapies based on GLP-1. Semaglutide, developed to achieve a longer half-life than liraglutide, enables once-weekly administration and induced clinically significant weight loss in randomized controlled clinical trials [22]. Recent randomized controlled trial studies have also proven that weekly semaglutide was superior compared to placebo in reducing cardiovascular risks for obese patients [23].
Meanwhile, dual and triple agonists targeting receptors beyond GLP-1R are continuously developed in preclinical models. In 2009 the first form of dual agonists was discovered by Day et al. [24], targeting GLP-1R and the glucagon receptor (GCGR). In 2013, Day et al. [25] also discovered dual agonists targeting both GLP-1 and GIP receptors (GIPR). In 2015, triple agonists simultaneously targeting the GLP-1R, GCGR, and GIPR were also developed, whose weight loss in mice exceeded those of the dual agonists [26]. Clinical trials for these coagonist incretin drugs are still continuously ongoing worldwide. Recently, the GIP/GLP-1R co-agonist tirzepatide has gone through several randomized controlled trials and was approved by the FDA for T2DM and obesity in 2022 and 2023, respectively. Once-weekly administered tirzepatide proved to be both noninferior and superior in reducing glycosylated hemoglobin levels in T2DM patients and demonstrated robust weight loss in obese patients [27,28]. The emergence of clinically effective dual and triple agonists holds promise for enhanced metabolic outcomes, although the precise mechanisms of drug actions remain to be fully elucidated.
GLP-1 has long been identified and validated as a biomarker peptide for satiation and satiety under physiological conditions [29,30]. Satiation refers to the process occurring ‘during’ a meal event that prompts eating termination and is associated with meal size, while satiety refers to the process occurring ‘after’ a meal event, inhibiting further eating until the next meal [30,31]. Satiation gradually rises during a meal event as hunger diminishes, peaking at meal termination. After the meal event, satiety gradually drops over time and hunger returns, starting a new cycle [32].
The satiety cascade, first proposed in the 1980s and modified throughout, indicates that satiation and satiety are governed by the interplay of metabolic, cognitive, and sensory factors, which are interconnected components [31,33,34]. Studies indicate that sensory and cognitive perceptions of food can indeed influence satiation by brain signaling, while the mechanism of this process is yet to be fully uncovered [35].
It is well known that peripheral injection of GLP-1R agonists interacts with the CNS [36,37]. Several human studies have proven that the actions of GLP-1R agonists in the brain are regulated by sensory and cognitive factors. We have previously investigated the acute effects of lixisenatide injection compared to saline in measurements of functional magnetic resonance imaging (fMRI) responses to visual food cues. In T2DM patients, lixisenatide reduced activation in brain areas including the hypothalamus, temporal lobar regions, lateral ventricle, and cortical areas in obese individuals in contrast to lean T2DM individuals, who showed increased activation in the areas [38].
fMRI studies on other GLP-1R agonists have also provided further evidence of their cognitive effects. In obese T2DM patients, exenatide reduced putamen activity to high-calorie food pictures, while enhancing putamen responses to low-calorie food pictures [39,40]. In a study conducted in obese male patients all exenatide responders, defined as those who consumed lower food calories by exenatide administration compared to placebo, exhibited significantly higher hypothalamic connectivity measured by food cue-paired fMRI compared with placebo injection [41]. Furthermore, liraglutide treatment for 10 days in obese T2DM patients attenuated insula and putamen fMRI responses to food pictures in fasted and post-meal conditions, compared with insulin glargine injection [42].
Interestingly, the effects of GLP-1R agonists have also been studied in non-obese patients without T2DM. In fasted healthy normal weight patients, GLP-1 (7-36) amide injection reduced fMRI activity induced by food images, in all brain areas studied (amygdala, insula, caudate, NAc, orbitofrontal cortex, and putamen), compared to saline injection [43].
Collectively, these results highlight the cognitive effects of GLP-1R agonists in response to food cues, implying that the effects of GLP-1R agonists on satiation and satiety extend to cognitive processes.
The hypothalamus is a well-established area of the brain known to regulate feeding behavior and energy metabolism, where disruption can lead to obesity [44]. The actions of agouti-related peptide (AgRP) neurons and pro-opiomelanocortin (POMC) neurons in the arcuate nucleus (ARC), the most renowned cell types of the hypothalamus governing homeostatic actions and inducing hunger, were previously understood to be mainly regulated by internal nutritional status or hormones such as leptin or ghrelin [45]. However, fiber photometry studies validated that ARCAgRP neural activity is inhibited within seconds after visual sensory cognition or the taste of food, in the preingestive stage [46-48]. Strikingly, the amount of reduction in ARCAgRP neural activity was modulated by food calories, and the learning of nutritional value in a single trial promptly suppressed ARCAgRP neural activity [49]. These results demonstrate the feed-forward nature of ARCAgRP neurons through a learned anticipation process. The sudden drop in signals at the moment of sensory detection or food cognition may reflect the abrupt recognition of an anticipated resolution of energy deficit, rather than being driven by ingestive or hormonal signals which typically act over minutes to hours [50]. Our previous research further corroborated this viewpoint by proving that ARCAgRP neural activity accounts for computational modeling of ‘need,’ defined as predicted deficiency, which is the sum of current deficiency and future predicted changes [51]. This definition of ‘need’ implies the preemptive update of deficiency prior to ingestion, which can indeed occur through sensory cognition and anticipation of the future and may be advantageous for survival [52].
In addition, the leptin-receptor-expressing (LepR) neural activity in the dorsomedial hypothalamus (DMH), which sends GABAergic inhibitory inputs to ARCAgRP neurons, is also known to be rapidly increased by sensory detection, the taste of food, or food cues [48,53,54]. DMHLepR neurons receive afferents from the lateral hypothalamus (LH)Vglut2 neurons, which are known to be necessary for the rapid responses to food cues [54]. Overall, these animal studies demonstrate the cognitive feed-forward hunger regulation mechanism that occurs in the hypothalamic region.
The hypothalamus can also be hypothesized to regulate satiation or satiety in a feed-forward manner. Indeed, in physiological conditions, plasma GLP-1 has been suggested to increase not only after meals, but also in the pre-ingestive, cephalic phase, with its peak observed about an hour prior to meal initiation in rats [55]. The following part of the review will discuss the hypothalamic actions regarding GLP-1, including its effects on satiation in cognitive stages.
Various studies have focused on the hypothalamus as a target of different types of GLP-1R agonists. A study using fluorescently labeled lipidated analogues of exendin-4 showed that they are distributed in the hypothalamus [56]. Fluorescently labeled liraglutide reaches hypothalamic regions such as the ARC, paraventricular nucleus (PVH), and supraoptic nucleus (SON) after peripheral administration, with GLP-1R undergoing internalization after binding to liraglutide [57]. GLP-1RA binding to GLP-1R is indeed mediated by internalization of GLP-1R and GLP-1RA, consistent with the fact that GLP-1R is classified as a class B G protein-coupled receptor (GPCR) [57,58]. Whole brain microscopy following the injection of fluorescently labeled semaglutide revealed that semaglutide is present in parts of the hypothalamus, including the DMH, ARC, PVH, medial mamillary nucleus, and SON [59].
The specific mechanism of how GLP-1RAs can cross the blood-brain barrier and reach the hypothalamus has been a matter of debate. Hypothalamic tanycytes are glial cells located along the walls and floor of the third ventricle, which are known to play neuroendocrine roles such as detecting and transporting nutrients and hormones like insulin and GLP-1, as well as regulating neuronal activity [60-62]. The role of hypothalamic tanycytes has been studied in different types of GLP-1RAs. These cells are actively involved in the transport of exendin-4 to the brain cerebrospinal fluid [63]. In addition, GLP-1R expressions are found in tanycytes, and it is known that tanycytic GLP-1R is required for liraglutide’s anti-obesity actions via its transport to hypothalamic cells [64]. Semaglutide can also bind to and activate tanycyte GLP-1Rs [59]. Further elucidation is needed about the function of tanycytes and tanycytic GLP-1R-dependent transport of GLP-1RAs.
The distribution of GLP-1R and the role of GLP-1R expressing neurons along with the effects of GLP-1RA in different areas of the hypothalamus have been thoroughly studied in various studies (Table 1, Fig. 1) [65-67].
Dorsomedial hypothalamus
GLP-1R are known to be expressed in the neurons of the DMH of mice and humans, and previous studies have emphasized its role in regulating metabolic responses by exendin-4 [68,69]. Direct injection of exendin-4 to the DMH reduced blood glucose levels, while liraglutide injection into the DMH did not alter 24-hour food intake or body weight [70,71]. Brain slice studies have shown that exendin-4 can increase the firing rates of DMH neurons, enhancing the neurons’ sensitivity to a high-fat diet (HFD) compared to a control diet [72]. Long-term ablation of DMH GLP-1R resulted in hyperphagia and obesity, and blunted exendin-4-induced anorexigenic metabolic effects [68,73].
The DMH GLP-1R expressing cells are known to be predominantly GABAergic, while a smaller percentage of the population do express vesicular glutamate transporter 2 (VGLUT2) mRNA [68]. Activated by GLP-1, DMH GLP-1R neurons are known to be crucial for glucose regulation, mediated by a cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) dependent pathway. These glucose-sensitive actions operate through downstream connections to the dorsal motor nucleus of the vagus (DMV) to the pancreas [70,73].
To further investigate the function of DMH GLP-1R neurons, our group conducted a translational study to elucidate their behavioral role. Optogenetic activation of the neurons triggered immediate meal termination and reduced bout duration and frequency, while neural inhibition prolonged bout duration and increased food intake, showing that the neurons are necessary and sufficient for satiation. Furthermore, calcium signal recording of DMH GLP-1R neurons highlighted that learned anticipation of food induces a significant increase in neural activity even before contact with food. In addition, exendin-4 injection enhanced calcium signals in DMH GLP-1R neurons in the pre-ingestive stage, and exendin-4-induced pre-ingestive satiation was mitigated by optogenetic inhibition of DMH GLP-1R neurons. These findings confirm that DMH GLP-1R neurons mediate pre-ingestive, cognitive satiation elicited by GLP-1R agonists, a phenomenon that we also observed in a human study [69]. The role of DMH GLP-1R neurons can be likened to stepping on a vehicle’s brake in response to a traffic signal—a learned anticipatory mechanism that prepares for meal termination independently of gut-derived feedback. This demonstrates a cognitive regulation of satiation, in which a feed-forward mechanism increases satiation before the initiation of a meal, eventually leading to meal termination (Fig. 2).
GLP-1R-LepR co-expressing neurons, localized primarily in the DMH in rodent and non-human primate models, mediate the suppression of food intake by liraglutide [74-76]. A recent study combining single-nucleus sequencing and spatial transcriptomics pinpointed the human hypothalamic GLP-1R-LepR co-expressing cluster, which strongly correlated with the mouse hypothalamic cluster that exhibits the second highest expression of GLP-1R in the hypothalamus [76]. Notably, GLP-1R-LepR co-expressing cells proved to be crucial targets of newly synthesized GLP-1/leptin dual agonists, which create a negative energy balance and assist the action of leptin [75].
As previously mentioned, DMH LepR neurons are known to be activated by sensory cue information and the taste of food [48,53,54]. Activation of DMH LepR neurons has been shown to be sufficient to inhibit food intake [53]. However, the authors concluded that DMH LepR neurons are not essential for satiation or satiety, as the study found no significant increase in food intake or body weight following inhibition or disruption of these neurons [54]. More recently, another study demonstrated that DMH LepR neurons respond to oro-sensory cues, acting as negative feedback signals to promote meal termination. Chemogenetic inhibition of DMH LepR neurons led to increased food intake by increasing the number of licking bouts without altering bout length, in contrast to the increased bout duration in DMH GLP-1R neurons, implying that DMH LepR neurons play a role in satiety [48]. Some of this discrepancy may result from differences in observed behavior (licking vs. eating), or from the complexities of semantic distinctions between terms such as satiation or satiety. Additionally, some discrepancies may be explained by our findings, which show that mRNA co-localization of LepR and GLP-1R varies depending on the anatomical location within the DMH, with a higher overlap percentage in the caudal part of the DMH than the rostral part [69]. Some studies have also found a higher density of GLP-1R-positive cells in the dorsal DMH (dDMH) than the ventral DMH (vDMH), and a higher density of GLP- 1-positive neuronal fibers in the vDMH than the dDMH [72]. Moreover, the LepR neurons specifically within the dDMH were recently studied for their role in activating brown adipose tissue (BAT) thermogenesis, while not altering food intake [77]. Further research is needed to clarify regional differences within the DMH in the expression of GLP-1R and LepR, their upstream and downstream circuits, and how they can contribute to satiation or satiety in different feeding phases. Furthermore, the role of DMH GLP-1R neurons or LepR neurons in sensory-specific satiety, a phenomenon in which repeated exposure to a particular taste reduces food intake, is an area for further study.
Arcuate nucleus
The ARC of the hypothalamus is one of the sites with abundant GLP-1R distribution [78]. Direct injection of 1 nmol GLP-1, exendin-4, or liraglutide to the ARC significantly suppressed food intake and body weight, with the effects observed over the 24 hours following injection [71,79,80].
It has been shown that GLP-1Rs are expressed in POMC neurons in the arcuate, while not in the neuropeptide Y (NPY)/AgRP neurons of the ARC [57,81]. Some studies have reported that knockdown of GLP-1R in ARC POMC neurons didn’t alter body weight or composition, food intake, and energy expenditure, nor did it significantly alter the reduction of food intake by intraperitoneal (i.p.) exendin-4 [80]. However, other studies have indicated that POMC GLP-1R knockdown can increase HFD-induced weight gain, and ARC GLP-1R blockade by GLP-1R antagonists attenuated the anorexic effects of liraglutide [57]. This discrepancy may be due to the different drug types, or intervention of other pathways or compensatory mechanisms after disruption of GLP-1R. Further studies are needed for elucidation.
Exendin-4, liraglutide and semaglutide can directly or indirectly stimulate POMC neurons in the ARC, while indirectly inhibiting NPY/AgRP neurons via presynaptic GABAergic neurons [82-84]. While previous fiber photometry results showed that administration of GLP-1 or liraglutide did not significantly alter ARC AgRP/NPY neuron calcium signals post 30-minute injection, after 2 hours post-injection, the rapid suppression of AgRP calcium signals after cognition of food was indeed attenuated by liraglutide [49,82,85]. Our patch-clamp electrophysiology results corroborate these findings, demonstrating that GLP-1RAs directly activate DMH GLP-1R neurons, which in turn send inhibitory projections to ARC AgRP/NPY neurons [69].
Chemogenetic activation of GLP-1R neurons in the ARC, particularly neurons co-expressing POMC and GLP-1R in male mice, significantly suppressed feeding [86,87]. In addition to POMC, other markers in the ARC that potentially represent targets for GLP-1RAs are currently being studied. ARC GABAergic neurons that express thyrotropin-releasing hormone (TRH), approximately 60% of which expressed GLP-1R, were found to have GABAergic afferents directly targeting AgRP neurons. ARC TRH neurons were directly activated by liraglutide and were shown to be necessary for its anorectic actions [88].
Paraventricular nucleus
The PVH is also a region known to have a dense population of GLP-1R in rodents [89]. In situ hybridization (ISH) studies have revealed that GLP-1R mRNA is also expressed in the human brain PVH, with its expression significantly reduced in T2DM patients compared to controls, potentially contributing to homeostatic imbalance [90]. Direct injection of GLP-1, exendin-4, or liraglutide to the PVH indeed suppressed food intake [71,80].
Single minded-1 (Sim1) is a transcription factor expressed in most PVH cells, and PVH Sim1 neurons are known to play a role in regulating satiety [91,92]. Some studies have reported that knockdown of GLP-1R in Sim1 neurons did not affect body weight, body composition, or food intake, nor did it significantly change the anorexic effects induced by i.p. exendin-4 injection [80]. Similarly, the blockade of GLP-1R in the PVH with antagonists failed to attenuate liraglutide-induced weight loss [57]. Yet GLP-1R knockdown in Sim1 neurons did result in a significant reduction in energy expenditure, in contrary to results from whole-body or hypothalamic GLP-1R knockout models. This suggests the presence of compensatory mechanisms involving other hypothalamic or extra-hypothalamic regions [80,93]. In addition, PVH GLP-1R knockout in juvenile mice led to increased adult body weight and food intake with no changes in energy expenditure, suggesting potential compensatory adaptation during development [94].
A study has shown that anticipation of food in the pre-ingestive stage, without ingestion and triggered solely by sensory detection of food, led to a rapid increase in calcium activity of PVH GLP-1R neurons. Chemogenetic activation and inhibition of PVH GLP-1R neurons were shown to reduce and increase food intake, respectively [95]. However, liraglutide did not directly activate Fos expression in the PVH, suggesting that the effects of GLP-1RA on this region may be indirect [95,96].
Other hypothalamic regions
The LH GLP-1R is also considered one of the hypothalamic targets in studies investigating central GLP-1R signaling. Injection of exendin-4 into the LH reduced lever-pressing behavior, while injection of exendin-9, a GLP-1R antagonist, increased food reinforcement. Additionally, liraglutide injection to the LH lowered food intake and body weight [71]. Interestingly, chronic LH GLP-1R knockdown resulted in increased nucleus accumbens (NAc) GLP-1R expression in male mice, which may be associated with compensation related to reward [97].
The ventromedial hypothalamus (VMH), a classically known satiety center, is also a brain area that has been studied [98]. Liraglutide injection into the VMH of mice induced weight loss and increased thermogenetic activity, as evidenced by elevated uncoupling protein 1 expression in both BAT and white adipose tissue, with adenosine monophosphate-activated protein kinase (AMPK) signaling in the VMH required as a mediator [71]. Similarly, exendin-4 injection into the VMH of mice also reduced food intake and body weight, but, interestingly, decreased energy expenditure—unlike liraglutide injection—despite both acting via AMPK inhibition [99]. Furthermore, VMH GLP-1R deletion did not significantly affect the anorexigenic effects of systemically administered exendin-4, suggesting that the VMH may not be a critical site for mediating GLP- 1RA driven appetite suppression [99]. Notably, GLP-1R has been reported to be absent in the VMH based on rat immunochemistry and non-human primates ISH studies, warranting caution when extrapolating these findings across species [66,67].
Non-hypothalamic regions
A variety of non-hypothalamic regions are also studied for the neural circuitry targets of GLP-1RA actions. Nausea and vomiting are among the most common side effects of GLP-1R agonists. The area postrema (AP) is a circumventricular organ of the hindbrain with a leaky blood-brain barrier, previously identified as a region associated with nausea and vomiting [100,101]. GLP-1R is expressed in the AP and nucleus of the solitary tract (NTS) located in the hindbrain, and c-Fos expression after GLP-1RA injection was apparent in hindbrain GLP-1R cells, with further studies demonstrating these cells’ role in mediating GLP-1RA actions [59,96,102,103].
A recent study distinguished the hindbrain neural circuits that mediate satiety and aversion. NTS GLP-1R neurons, sensitive to nutritive stimuli, induced satiety without aversion, whereas AP GLP-1R neurons elicited strong nausea-like behavior in mice. Ablation of AP GLP-1R neurons decreased aversion while maintaining appetite suppression, indicating the possibility of future incretin drugs that can reduce nausea by curbing the aversion pathway [104].
The lateral septum (LS) is also one of the regions known to have high expression of GLP-1R [105,106]. Direct injection of GLP-1RA into the LS reduced overnight chow and HFD intake [107]. Moreover, stimulation of LS GLP-1R neurons reduced food intake, both in an acute and chronic manner, while inhibition increased food intake [108,109]. Systemic liraglutide administration activated LS GLP-1R neurons, and the knockdown of GLP-1Rs in the LS mitigated the anti-obesity effects of liraglutide. Interestingly, unlike DMH GLP-1R neurons, calcium signals of LS GLP-1R neurons decreased at initiation of food consumption [109]. Recent studies have also shown that LS GLP-1R can play a role in mitigating cocaine-induced locomotion and cocaine conditioned place preference [110]. Indeed, i.p. injection of exenatide dampened cocaine-driven septal dopamine elevation, mediated by postsynaptic GLP-1R signaling that retrogradely regulates presynaptic dopamine transporter function [106].
Ongoing studies imply that the usage of GLP-1R agonists can be expanded to other areas, beyond their conventional roles in controlling food intake or energy balance. GLP-1RAs are being studied for their significant associations with reduced risks of various cardiovascular diseases, neuropsychiatric or substance use disorders, infectious illnesses and respiratory conditions, indicating their potential for broader therapeutic applications [111]. Notably, GLP-1R agonists have recently been suggested to have a role in mitigating reward-seeking. A vast number of preclinical and clinical studies have provided evidence for the potential role of GLP-1R agonists in modulating reward-seeking of food, alcohol, cocaine, and amphetamine, offering the possibility of a novel therapeutic alternative for treating addiction [112]. Randomized controlled clinical trials investigating the use of semaglutide in patients with alcohol use disorders (AUD) and comorbid obesity are currently underway, based on the potential of GLP-1R agonists for AUD treatment [113]. More studies are needed to clarify the underlying mechanism of drug and GLP-1 actions on reward circuits.
Recently, novel drugs combining GLP-1R agonism with other options, beyond the forementioned dual or triple agonist drugs, are being developed to maximize efficacy and minimize adverse effects in obesity and diabetes treatment. For instance, combinations of GLP-1R agonists with amylin analogues or peptide YY (PYY) analogues are currently undergoing clinical trials [114]. In addition, in preclinical stages, a combination of GLP-1R agonism with N-methyl-D-aspartate (NMDA) receptor antagonism (GLP-1-MK-801) has been developed [115]. Furthermore, small molecule oral agonists such as danuglipron are also under development, highlighting the potential for improved patient compliance through non-injectable delivery forms [116].
Several challenges remain to be addressed. The presence of non-responders to GLP-1R therapy continues to complicate treatment outcomes, underscoring the need for a deeper understanding of individual variability in response [117,118]. Additionally, the issues of weight regain and altered metabolic outcomes following drug discontinuation emphasize the importance of long-term studies investigating the impacts of the hormonal and CNS on weight regain to optimize safety and efficacy [119,120].
In conclusion, the DMH is a key target of GLP-1RAs, mediating pre-ingestive satiation, which is a cognitive process. GLP-1R neurons in the DMH and other hypothalamic nuclei regulate multiple aspects of eating behavior, presenting potential therapeutic targets for GLP-1R-based therapies and other drug candidates.

CONFLICTS OF INTEREST

No potential conflict of interest relevant to this article was reported.

FUNDING

None

Acknowledgements
We thank our lab members for support and discussion.
Fig. 1.
Overview of hypothalamic glucagon-like peptide-1 receptor (GLP-1R) neurons. PVH, paraventricular hypothalamic nucleus; Oxt, oxytocin-expressing; Crh, corticotropin-releasing hormone-expressing; MC4R, melanocortin 4 receptor-expressing; DMH, dorsomedial hypothalamus; LepR, leptin-receptor-expressing; LH, lateral hypothalamus; dDMH, dorsal DMH; vDMH, ventral DMH; GLP-1, glucagon-like peptide-1; ARC, arcuate nucleus; AgRP, agouti-related peptide; POMC, pro-opiomelanocortin; TRH, thyrotropin-releasing hormone-expressing; VMH, ventromedial hypothalamus; i.p., intraperitoneal.
dmj-2025-0106f1.jpg
Fig. 2.
Feed-forward cognitive control of satiation. The role of dorsomedial hypothalamus (DMH) glucagon-like peptide-1 receptor (GLP-1R) neurons regulating cognitive satiation can be likened to stepping on a vehicle’s brake in response to traffic signals— a preemptive action taken before reaching the actual stop goal, reflecting a feed-forward control mechanism. These neurons project inhibitory signals to arcuate nucleus (ARC) agouti-related peptide (AgRP) neurons, ultimately contributing to meal termination. Notably, glucagon-like peptide-1 receptor agonists (GLP-1RAs) can potentiate the activity of DMH GLP-1R neurons.
dmj-2025-0106f2.jpg
dmj-2025-0106f3.jpg
Table 1.
Animal studies of GLP-1R, GLP-1R neurons, GLP-1RA effects on hypothalamus
Brain region Mice GLP-1R transgene [65]a Rat GLP-1R immunocytochemistry [66]a Monkey GLP-1R ISH [67]a Direct injection of GLP-1RA to brain region Disruption of GLP-1R in the area GLP-1RA i.p. injection +, Disruption of GLP-1R in the area Calcium signal of brain region GLP-1R neurons Neuromodulation of brain region GLP-1R neurons
GLP-1RA i.p. injection effects on brain region GLP-1R neurons
Activation Inhibition
DMH ++ +++ ++ (Exendin-4) (DMH glp1r KD or ablation) (i.p. Exendin-4+glp1r KD) Neural activity ↑at pre-ingestive, or ingestive stages Bout duration↓ Bout duration↑ (Exendin-4)
Blood glucose level ↓ B.W.↑ |Δ Food intake| ↓ Number of bout↓ Food intake↑ Potentiate DMH GLP-1R neurons’ calcium signals
(Liraglutide) Food intake↑ |Δ B.W. | ↓ Latency to ingestion termination ↓
B.W. ~ Plasma TG↑ |Δ E.E. |↓
Food intake ~ E.E. ↓ |Δ RER |↓
UCP1↓
Feed pattern disruption
ARC +++ ++++ ++++ (GLP-1, exendin-4, liraglutide) (POMC glp1r KD) (i.p. Exendin-4+POMC glp1r KD) Food intake ↓ (Liraglutide, semaglutide)
B.W. ↓ B.W. ~ |Δ Food intake| ~ Increase excitatory tone to POMC neurons
Food intake↓ HFD-induced weight gain ↑ (i.p. Liraglutide+ARC glp1r antagonist)
Food intake ~ |Δ Food intake| ↓
E.E. ~
PVH +++ +++ +++ (GLP-1, exendin-4, liraglutide) (Sim1 glp1r KD) (i.p. Exendin-4+Sim1 glp1r KD) Neural activity ↑at pre-ingestive, or ingestive stages Food intake ↓ Food intake ↑
B.W. ↓ B.W. ~ |Δ Food intake| ~
Food intake↓ HFD-induced weight gain ~ (i.p. Liraglutide+PVH glp1r antagonist)
Food intake ~ |Δ Food intake| ~
E.E. ↓
(Juvenile PVH glp1r KO)
B.W.↑
Food intake↑
E.E. ~
(PVH glp1r antagonist)
B.W.↑
LH ++ ++ ++ (Exendin-4) NAc GLP-1R expression ↑
Lever-pressing ↓
(Liraglutide)
B.W. ↓
Food intake↓
VMH +++ - - (Exendin-4) (VMH glp1r KD) (Exendin-4) - - - -
B.W. ↓ Food intake ~ |Δ Food intake| ~
Food intake↓ B.W. ~ (Liraglutide)
E.E. ↓ E.E. ~ |Δ B.W. | ~
(Liraglutide)
B.W. ↓
Food intake ~

GLP-1R, glucagon-like peptide-1 receptor; GLP-1RA, glucagon-like peptide-1 receptor agonist; ISH, in situ hybridization; DMH, dorsomedial hypothalamus; B.W., body weight; KD, knockdown; TG, triglyceride; E.E., energy expenditure; UCP1, uncoupling protein 1; i.p., intraperitoneal; RER, resting energy requirement; ARC, arcuate nucleus; POMC, pro-opiomelanocortin; HFD, high-fat diet; PVH, paraventricular hypothalamic nucleus; KO, knockout; Sim1, single minded-1; LH, lateral hypothalamus; NAc, nucleus accumbens; VMH, ventromedial hypothalamus.

a GLP-1R expression detected by transgene expression in mice [65], density of GLP-1R-immunoreactive fibers in each brain region determined by rat immunocytochemistry [66], GLP-1R expression determined by ISH in non-human primates [67].

  • 1. Lim GE, Brubaker PL. Glucagon-like peptide 1 secretion by the L-cell: the view from within. Diabetes 2006;55(Supplement 2):S70-7.
  • 2. Bayliss WM, Starling EH. The mechanism of pancreatic secretion. J Physiol 1902;28:325-53.ArticlePubMedPMC
  • 3. Elrick H, Stimmler L, Hlad CJ Jr, Arai Y. Plasma insulin response to oral and intravenous glucose administration. J Clin Endocrinol Metab 1964;24:1076-82.PubMed
  • 4. Mcintyre N, Holdsworth CD, Turner DS. New interpretation of oral glucose tolerance. Lancet 1964;2:20-1.Article
  • 5. Dupre J, Ross SA, Watson D, Brown JC. Stimulation of insulin secretion by gastric inhibitory polypeptide in man. J Clin Endocrinol Metab 1973;37:826-8.PubMed
  • 6. Lund PK, Goodman RH, Dee PC, Habener JF. Pancreatic preproglucagon cDNA contains two glucagon-related coding sequences arranged in tandem. Proc Natl Acad Sci U S A 1982;79:345-9.ArticlePubMedPMC
  • 7. Lund PK. The discovery of glucagon-like peptide 1. Regul Pept 2005;128:93-6.ArticlePubMed
  • 8. Bell GI, Santerre RF, Mullenbach GT. Hamster preproglucagon contains the sequence of glucagon and two related peptides. Nature 1983;302:716-8.ArticlePubMedPDF
  • 9. Mojsov S, Weir GC, Habener JF. Insulinotropin: glucagon-like peptide I (7-37) co-encoded in the glucagon gene is a potent stimulator of insulin release in the perfused rat pancreas. J Clin Invest 1987;79:616-9.ArticlePubMedPMC
  • 10. Holst JJ, Orskov C, Nielsen OV, Schwartz TW. Truncated glucagon-like peptide I, an insulin-releasing hormone from the distal gut. FEBS Lett 1987;211:169-74.ArticlePubMedPDF
  • 11. Kreymann B, Williams G, Ghatei MA, Bloom SR. Glucagon-like peptide-1 7-36: a physiological incretin in man. Lancet 1987;5;2:1300-4.ArticlePubMedPMC
  • 12. Nathan DM, Schreiber E, Fogel H, Mojsov S, Habener JF. Insulinotropic action of glucagonlike peptide-I-(7-37) in diabetic and nondiabetic subjects. Diabetes Care 1992;15:270-6.ArticlePubMedPDF
  • 13. Sharma D, Verma S, Vaidya S, Kalia K, Tiwari V. Recent updates on GLP-1 agonists: current advancements & challenges. Biomed Pharmacother 2018;108:952-62.ArticlePubMed
  • 14. Eng J, Kleinman WA, Singh L, Singh G, Raufman JP. Isolation and characterization of exendin-4, an exendin-3 analogue, from Heloderma suspectum venom: further evidence for an exendin receptor on dispersed acini from guinea pig pancreas. J Biol Chem 1992;267:7402-5.ArticlePubMed
  • 15. Christel CM, DeNardo DF, Secor SM. Metabolic and digestive response to food ingestion in a binge-feeding lizard, the Gila monster (Heloderma suspectum). J Exp Biol 2007;210(Pt 19):3430-9.ArticlePubMedPDF
  • 16. Yap MKK, Misuan N. Exendin-4 from Heloderma suspectum venom: from discovery to its latest application as type II diabetes combatant. Basic Clin Pharmacol Toxicol 2019;124:513-27.PubMed
  • 17. Goke R, Fehmann HC, Linn T, Schmidt H, Krause M, Eng J, et al. 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;268:19650-5.ArticlePubMed
  • 18. Kurtzhals P, Havelund S, Jonassen I, Kiehr B, Larsen UD, Ribel U, et al. Albumin binding of insulins acylated with fatty acids: characterization of the ligand-protein interaction and correlation between binding affinity and timing of the insulin effect in vivo. Biochem J 1995;312(Pt 3):725-31.ArticlePubMedPMCPDF
  • 19. Astrup A, Rossner S, Van Gaal L, Rissanen A, Niskanen L, Al Hakim M, et al. Effects of liraglutide in the treatment of obesity: a randomised, double-blind, placebo-controlled study. Lancet 2009;374:1606-16.ArticlePubMed
  • 20. Pi-Sunyer X, Astrup A, Fujioka K, Greenway F, Halpern A, Krempf M, et al. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med 2015;373:11-22.ArticlePubMed
  • 21. Park JS, Kwon J, Choi HJ, Lee C. Clinical effectiveness of liraglutide on weight loss in South Koreans: first real-world retrospective data on Saxenda in Asia. Medicine (Baltimore) 2021;100:e23780.PubMedPMC
  • 22. Wilding JP, Batterham RL, Calanna S, Davies M, Van Gaal LF, Lingvay I, et al. Once-weekly semaglutide in adults with overweight or obesity. N Engl J Med 2021;384:989-1002.ArticlePubMed
  • 23. Lincoff AM, Brown-Frandsen K, Colhoun HM, Deanfield J, Emerson SS, Esbjerg S, et al. Semaglutide and cardiovascular outcomes in obesity without diabetes. N Engl J Med 2023;389:2221-32.ArticlePubMed
  • 24. Day JW, Ottaway N, Patterson JT, Gelfanov V, Smiley D, Gidda J, et al. A new glucagon and GLP-1 co-agonist eliminates obesity in rodents. Nat Chem Biol 2009;5:749-57.ArticlePubMedPDF
  • 25. Finan B, Ma T, Ottaway N, Muller TD, Habegger KM, Heppner KM, et al. Unimolecular dual incretins maximize metabolic benefits in rodents, monkeys, and humans. Sci Transl Med 2013;5:209ra151.ArticlePubMed
  • 26. Finan B, Yang B, Ottaway N, Smiley DL, Ma T, Clemmensen C, et al. A rationally designed monomeric peptide triagonist corrects obesity and diabetes in rodents. Nat Med 2015;21:27-36.PubMed
  • 27. Frias JP, Davies MJ, Rosenstock J, Perez Manghi FC, Fernandez Lando L, Bergman BK, et al. Tirzepatide versus semaglutide once weekly in patients with type 2 diabetes. N Engl J Med 2021;385:503-15.ArticlePubMed
  • 28. Jastreboff AM, Aronne LJ, Ahmad NN, Wharton S, Connery L, Alves B, et al. Tirzepatide once weekly for the treatment of obesity. N Engl J Med 2022;387:205-16.ArticlePubMed
  • 29. de Graaf C, Blom WA, Smeets PA, Stafleu A, Hendriks HF. Biomarkers of satiation and satiety. Am J Clin Nutr 2004;79:946-61.ArticlePubMed
  • 30. Tremblay A, Bellisle F. Nutrients, satiety, and control of energy intake. Appl Physiol Nutr Metab 2015;40:971-9.ArticlePubMed
  • 31. Blundell J, de Graaf C, Hulshof T, Jebb S, Livingstone B, Lluch A, et al. Appetite control: methodological aspects of the evaluation of foods. Obes Rev 2010;11:251-70.ArticlePubMedPMC
  • 32. Tack J, Verbeure W, Mori H, Schol J, Van den Houte K, Huang IH, et al. The gastrointestinal tract in hunger and satiety signalling. United European Gastroenterol J 2021;9:727-34.ArticlePubMedPMCPDF
  • 33. Mela DJ. Eating for pleasure or just wanting to eat? Reconsidering sensory hedonic responses as a driver of obesity. Appetite 2006;47:10-7.ArticlePubMed
  • 34. Colms J, Booth DA, Pangborn RM, Raunhardt O. Food acceptance and nutrition. London: Academic Press; 1987. Chapter, Evaluating the satiating power of foods: implications for acceptance and consumption; p205-19.
  • 35. Lasschuijt MP, de Graaf K, Mars M. Effects of oro-sensory exposure on satiation and underlying neurophysiological mechanisms: what do we know so far? Nutrients 2021;13:1391.ArticlePubMedPMC
  • 36. Kastin AJ, Akerstrom V. Entry of exendin-4 into brain is rapid but may be limited at high doses. Int J Obes Relat Metab Disord 2003;27:313-8.ArticlePubMedPDF
  • 37. Kastin AJ, Akerstrom V, Pan W. Interactions of glucagon-like peptide-1 (GLP-1) with the blood-brain barrier. J Mol Neurosci 2002;18:7-14.ArticlePubMed
  • 38. Bae JH, Choi HJ, Cho KIK, Kim LK, Kwon JS, Cho YM. Glucagon-like peptide-1 receptor agonist differentially affects brain activation in response to visual food cues in lean and obese individuals with type 2 diabetes mellitus. Diabetes Metab J 2020;44:248-59.ArticlePubMedPDF
  • 39. van Ruiten CC, Veltman DJ, Nieuwdorp M, IJzerman RG. Brain activation in response to low-calorie food pictures: an explorative analysis of a randomized trial with dapagliflozin and exenatide. Front Endocrinol (Lausanne) 2022;13:863592.PubMedPMC
  • 40. van Ruiten CC, Veltman DJ, Schrantee A, van Bloemendaal L, Barkhof F, Kramer MH, et al. Effects of dapagliflozin and combination therapy with exenatide on food-cue induced brain activation in patients with type 2 diabetes. J Clin Endocrinol Metab 2022;107:e2590. -9.ArticlePubMedPMCPDF
  • 41. Schlogl H, Kabisch S, Horstmann A, Lohmann G, Muller K, Lepsien J, et al. Exenatide-induced reduction in energy intake is associated with increase in hypothalamic connectivity. Diabetes Care 2013;36:1933-40.ArticlePubMedPMCPDF
  • 42. Ten Kulve JS, Veltman DJ, van Bloemendaal L, Barkhof F, Drent ML, Diamant M, et al. Liraglutide reduces CNS activation in response to visual food cues only after short-term treatment in patients with type 2 diabetes. Diabetes Care 2016;39:214-21.ArticlePubMedPDF
  • 43. De Silva A, Salem V, Long CJ, Makwana A, Newbould RD, Rabiner EA, et al. The gut hormones PYY 3-36 and GLP-1 7-36 amide reduce food intake and modulate brain activity in appetite centers in humans. Cell Metab 2011;14:700-6.ArticlePubMedPMC
  • 44. Timper K, Bruning JC. Hypothalamic circuits regulating appetite and energy homeostasis: pathways to obesity. Dis Model Mech 2017;10:679-89.ArticlePubMedPMCPDF
  • 45. Chen Y, Knight ZA. Making sense of the sensory regulation of hunger neurons. Bioessays 2016;38:316-24.ArticlePubMedPMCPDF
  • 46. Betley JN, Xu S, Cao ZF, Gong R, Magnus CJ, Yu Y, et al. Neurons for hunger and thirst transmit a negative-valence teaching signal. Nature 2015;521:180-5.ArticlePubMedPMCPDF
  • 47. Chen Y, Lin YC, Kuo TW, Knight ZA. Sensory detection of food rapidly modulates arcuate feeding circuits. Cell 2015;160:829-41.ArticlePubMedPMC
  • 48. Aitken TJ, Liu Z, Ly T, Shehata S, Sivakumar N, La Santa Medina N, et al. Negative feedback control of hypothalamic feeding circuits by the taste of food. Neuron 2024;112:3354-70.ArticlePubMed
  • 49. Su Z, Alhadeff AL, Betley JN. Nutritive, post-ingestive signals are the primary regulators of AgRP neuron activity. Cell Rep 2017;21:2724-36.ArticlePubMedPMC
  • 50. Andermann ML, Lowell BB. Toward a wiring diagram understanding of appetite control. Neuron 2017;95:757-78.ArticlePubMedPMC
  • 51. Kim KS, Lee YH, Yun JW, Kim YB, Song HY, Park JS, et al. A normative framework dissociates need and motivation in hypothalamic neurons. Sci Adv 2024;10:eado1820.ArticlePubMedPMC
  • 52. Kim YB, Lee YH, Park SJ, Choi HJ. A unified theoretical framework underlying the regulation of motivated behavior. Bioessays 2024;46:e2400016.ArticlePubMed
  • 53. Garfield AS, Shah BP, Burgess CR, Li MM, Li C, Steger JS, et al. Dynamic GABAergic afferent modulation of AgRP neurons. Nat Neurosci 2016;19:1628-35.ArticlePubMedPMCPDF
  • 54. Berrios J, Li C, Madara JC, Garfield AS, Steger JS, Krashes MJ, et al. Food cue regulation of AGRP hunger neurons guides learning. Nature 2021;595:695-700.ArticlePubMedPMCPDF
  • 55. Vahl TP, Drazen DL, Seeley RJ, D’Alessio DA, Woods SC. Meal-anticipatory glucagon-like peptide-1 secretion in rats. Endocrinology 2010;151:569-75.ArticlePubMed
  • 56. Skovbjerg G, Roostalu U, Salinas CG, Skytte JL, Perens J, Clemmensen C, et al. Uncovering CNS access of lipidated exendin-4 analogues by quantitative whole-brain 3D light sheet imaging. Neuropharmacology 2023;238:109637.ArticlePubMed
  • 57. Secher A, Jelsing J, Baquero AF, Hecksher-Sorensen J, Cowley MA, Dalboge LS, et al. The arcuate nucleus mediates GLP-1 receptor agonist liraglutide-dependent weight loss. J Clin Invest 2014;124:4473-88.ArticlePubMedPMC
  • 58. Graaf Cd, Donnelly D, Wootten D, Lau J, Sexton PM, Miller LJ, et al. Glucagon-like peptide-1 and its class B G proteincoupled receptors: a long march to therapeutic successes. Pharmacol Rev 2016;68:954-1013.ArticlePubMedPMC
  • 59. Gabery S, Salinas CG, Paulsen SJ, Ahnfelt-Ronne J, Alanentalo T, Baquero AF, et al. Semaglutide lowers body weight in rodents via distributed neural pathways. JCI Insight 2020;5:e133429.ArticlePubMedPMC
  • 60. Goodman T, Hajihosseini MK. Hypothalamic tanycytes-masters and servants of metabolic, neuroendocrine, and neurogenic functions. Front Neurosci 2015;9:387.ArticlePubMedPMC
  • 61. Dali R, Estrada-Meza J, Langlet F. Tanycyte, the neuron whisperer. Physiol Behav 2023;263:114108.ArticlePubMed
  • 62. Bruning JC, Fenselau H. Integrative neurocircuits that control metabolism and food intake. Science 2023;381:eabl7398.PubMed
  • 63. Bakker W, Imbernon M, Salinas CG, Moro Chao DH, Hassouna R, Morel C, et al. Acute changes in systemic glycemia gate access and action of GLP-1R agonist on brain structures controlling energy homeostasis. Cell Rep 2022;41:111698.ArticlePubMedPMC
  • 64. Imbernon M, Saponaro C, Helms HC, Duquenne M, Fernandois D, Deligia E, et al. Tanycytes control hypothalamic liraglutide uptake and its anti-obesity actions. Cell Metab 2022;34:1054-63.ArticlePubMedPMC
  • 65. Cork SC, Richards JE, Holt MK, Gribble FM, Reimann F, Trapp S. Distribution and characterisation of Glucagon-like peptide-1 receptor expressing cells in the mouse brain. Mol Metab 2015;4:718-31.ArticlePubMedPMC
  • 66. Farkas E, Szilvasy-Szabo A, Ruska Y, Sinko R, Rasch MG, Egebjerg T, et al. Distribution and ultrastructural localization of the glucagon-like peptide-1 receptor (GLP-1R) in the rat brain. Brain Struct Funct 2021;226:225-45.ArticlePubMedPDF
  • 67. Heppner KM, Kirigiti M, Secher A, Paulsen SJ, Buckingham R, Pyke C, et al. Expression and distribution of glucagon-like peptide-1 receptor mRNA, protein and binding in the male nonhuman primate (Macaca mulatta) brain. Endocrinology 2015;156:255-67.ArticlePubMed
  • 68. Lee SJ, Sanchez-Watts G, Krieger JP, Pignalosa A, Norell PN, Cortella A, et al. Loss of dorsomedial hypothalamic GLP-1 signaling reduces BAT thermogenesis and increases adiposity. Mol Metab 2018;11:33-46.ArticlePubMedPMC
  • 69. Kim KS, Park JS, Hwang E, Park MJ, Shin HY, Lee YH, et al. GLP-1 increases preingestive satiation via hypothalamic circuits in mice and humans. Science 2024;385:438-46.ArticlePubMedPMC
  • 70. Huang Z, Liu L, Zhang J, Conde K, Phansalkar J, Li Z, et al. Glucose-sensing glucagon-like peptide-1 receptor neurons in the dorsomedial hypothalamus regulate glucose metabolism. Sci Adv 2022;8:eabn5345.ArticlePubMedPMC
  • 71. Beiroa D, Imbernon M, Gallego R, Senra A, Herranz D, Villarroya F, et al. GLP-1 agonism stimulates brown adipose tissue thermogenesis and browning through hypothalamic AMPK. Diabetes 2014;63:3346-58.ArticlePubMedPDF
  • 72. Sanetra AM, Palus-Chramiec K, Chrobok L, Jeczmien-Lazur JS, Klich JD, Lewandowski MH. Proglucagon signalling in the rat Dorsomedial Hypothalamus: physiology and high-fat dietmediated alterations. Mol Cell Neurosci 2023;126:103873.ArticlePubMed
  • 73. Maejima Y, Yokota S, Shimizu M, Horita S, Kobayashi D, Hazama A, et al. The deletion of glucagon-like peptide-1 receptors expressing neurons in the dorsomedial hypothalamic nucleus disrupts the diurnal feeding pattern and induces hyperphagia and obesity. Nutr Metab (Lond) 2021;18:58.ArticlePubMedPMCPDF
  • 74. Rupp AC, Tomlinson AJ, Affinati AH, Yacawych WT, Duensing AM, True C, et al. Suppression of food intake by Glp1r/Lepr- coexpressing neurons prevents obesity in mouse models. J Clin Invest 2023;133:e157515.ArticlePubMedPMC
  • 75. Polex-Wolf J, Deibler K, Hogendorf WF, Bau S, Glendorf T, Stidsen CE, et al. Glp1r-Lepr coexpressing neurons modulate the suppression of food intake and body weight by a GLP-1/ leptin dual agonist. Sci Transl Med 2024;16:eadk4908.ArticlePubMed
  • 76. Tadross JA, Steuernagel L, Dowsett GK, Kentistou KA, Lundh S, Porniece M, et al. A comprehensive spatio-cellular map of the human hypothalamus. Nature 2025;639:708-16.ArticlePubMedPMCPDF
  • 77. Francois M, Kaiser L, He Y, Xu Y, Salbaum JM, Yu S, et al. Leptin receptor neurons in the dorsomedial hypothalamus require distinct neuronal subsets for thermogenesis and weight loss. Metabolism 2025;163:156100.ArticlePubMed
  • 78. Ast J, Arvaniti A, Fine NH, Nasteska D, Ashford FB, Stamataki Z, et al. Super-resolution microscopy compatible fluorescent probes reveal endogenous glucagon-like peptide-1 receptor distribution and dynamics. Nat Commun 2020;11:467.ArticlePubMedPMCPDF
  • 79. NamKoong C, Kim MS, Jang BT, Lee YH, Cho YM, Choi HJ. Central administration of GLP-1 and GIP decreases feeding in mice. Biochem Biophys Res Commun 2017;490:247-52.ArticlePubMed
  • 80. Burmeister MA, Ayala JE, Smouse H, Landivar-Rocha A, Brown JD, Drucker DJ, et al. The hypothalamic glucagon-like peptide 1 receptor is sufficient but not necessary for the regulation of energy balance and glucose homeostasis in mice. Diabetes 2017;66:372-84.ArticlePubMedPDF
  • 81. Sandoval DA, Bagnol D, Woods SC, D’Alessio DA, Seeley RJ. Arcuate glucagon-like peptide 1 receptors regulate glucose homeostasis but not food intake. Diabetes 2008;57:2046-54.ArticlePubMedPMCPDF
  • 82. Dong Y, Carty J, Goldstein N, He Z, Hwang E, Chau D, et al. Time and metabolic state-dependent effects of GLP-1R agonists on NPY/AgRP and POMC neuronal activity in vivo. Mol Metab 2021;54:101352.ArticlePubMedPMC
  • 83. He Z, Gao Y, Lieu L, Afrin S, Cao J, Michael NJ, et al. Direct and indirect effects of liraglutide on hypothalamic POMC and NPY/AgRP neurons: implications for energy balance and glucose control. Mol Metab 2019;28:120-34.ArticlePubMedPMC
  • 84. Peterfi Z, Szilvasy-Szabo A, Farkas E, Ruska Y, Pyke C, Knudsen LB, et al. Glucagon-like peptide-1 regulates the proopiomelanocortin neurons of the arcuate nucleus both directly and indirectly via presynaptic action. Neuroendocrinology 2021;111:986-97.ArticlePubMedPDF
  • 85. Beutler LR, Chen Y, Ahn JS, Lin YC, Essner RA, Knight ZA. Dynamics of gut-brain communication underlying hunger. Neuron 2017;96:461-75.ArticlePubMedPMC
  • 86. Singh I, Wang L, Xia B, Liu J, Tahiri A, El Ouaamari A, et al. Activation of arcuate nucleus glucagon-like peptide-1 receptor-expressing neurons suppresses food intake. Cell Biosci 2022;12:178.ArticlePubMedPMCPDF
  • 87. Biglari N, Gaziano I, Schumacher J, Radermacher J, Paeger L, Klemm P, et al. Functionally distinct POMC-expressing neuron subpopulations in hypothalamus revealed by intersectional targeting. Nat Neurosci 2021;24:913-29.ArticlePubMedPMCPDF
  • 88. Webster AN, Becker JJ, Li C, Schwalbe DC, Kerspern D, Karolczak EO, et al. Molecular connectomics reveals a glucagon-like peptide 1-sensitive neural circuit for satiety. Nat Metab 2024;6:2354-73.ArticlePubMedPDF
  • 89. Xu XY, Wang JX, Chen JL, Dai M, Wang YM, Chen Q, et al. GLP-1 in the hypothalamic paraventricular nucleus promotes sympathetic activation and hypertension. J Neurosci 2024;44:e2032232024.ArticlePubMedPMC
  • 90. Ten Kulve JS, van Bloemendaal L, Balesar R, IJzerman RG, Swaab DF, Diamant M, et al. Decreased hypothalamic glucagon- like peptide-1 receptor expression in type 2 diabetes patients. J Clin Endocrinol Metab 2016;101:2122-9.ArticlePubMed
  • 91. Holder JL Jr, Butte NF, Zinn AR. Profound obesity associated with a balanced translocation that disrupts the SIM1 gene. Hum Mol Genet 2000;9:101-8.ArticlePubMed
  • 92. Li MM, Madara JC, Steger JS, Krashes MJ, Balthasar N, Campbell JN, et al. The paraventricular hypothalamus regulates satiety and prevents obesity via two genetically distinct circuits. Neuron 2019;102:653-67.ArticlePubMedPMC
  • 93. Hansotia T, Maida A, Flock G, Yamada Y, Tsukiyama K, Seino Y, et al. Extrapancreatic incretin receptors modulate glucose homeostasis, body weight, and energy expenditure. J Clin Invest 2007;117:143-52.ArticlePubMed
  • 94. Liu J, Conde K, Zhang P, Lilascharoen V, Xu Z, Lim BK, et al. Enhanced AMPA receptor trafficking mediates the anorexigenic effect of endogenous glucagon-like peptide-1 in the paraventricular hypothalamus. Neuron 2017;96:897-909.ArticlePubMedPMC
  • 95. Li C, Navarrete J, Liang-Guallpa J, Lu C, Funderburk SC, Chang RB, et al. Defined paraventricular hypothalamic populations exhibit differential responses to food contingent on caloric state. Cell Metab 2019;29:681-94.ArticlePubMed
  • 96. Adams JM, Pei H, Sandoval DA, Seeley RJ, Chang RB, Liberles SD, et al. Liraglutide modulates appetite and body weight through glucagon-like peptide 1 receptor-expressing glutamatergic neurons. Diabetes 2018;67:1538-48.ArticlePubMedPMCPDF
  • 97. Lopez-Ferreras L, Richard JE, Noble EE, Eerola K, Anderberg RH, Olandersson K, et al. Lateral hypothalamic GLP-1 receptors are critical for the control of food reinforcement, ingestive behavior and body weight. Mol Psychiatry 2018;23:1157-68.ArticlePubMedPDF
  • 98. Panksepp J. Is satiety mediated by the ventromedial hypothalamus? Physiol Behav 1971;7:381-4.Article
  • 99. Burmeister MA, Brown JD, Ayala JE, Stoffers DA, Sandoval DA, Seeley RJ, et al. The glucagon-like peptide-1 receptor in the ventromedial hypothalamus reduces short-term food intake in male mice by regulating nutrient sensor activity. Am J Physiol Endocrinol Metab 2017;313:E651-62.ArticlePubMedPMC
  • 100. Zhang C, Kaye JA, Cai Z, Wang Y, Prescott SL, Liberles SD. Area postrema cell types that mediate nausea-associated behaviors. Neuron 2021;109:461-72.ArticlePubMed
  • 101. Miyata S. Glial functions in the blood-brain communication at the circumventricular organs. Front Neurosci 2022;16:991779.ArticlePubMedPMC
  • 102. Ludwig MQ, Todorov PV, Egerod KL, Olson DP, Pers TH. Single-cell mapping of GLP-1 and GIP receptor expression in the dorsal vagal complex. Diabetes 2021;70:1945-55.ArticlePubMedPMCPDF
  • 103. Fortin SM, Lipsky RK, Lhamo R, Chen J, Kim E, Borner T, et al. GABA neurons in the nucleus tractus solitarius express GLP-1 receptors and mediate anorectic effects of liraglutide in rats. Sci Transl Med 2020;12:eaay8071.ArticlePubMedPMC
  • 104. Huang KP, Acosta AA, Ghidewon MY, McKnight AD, Almeida MS, Nyema NT, et al. Dissociable hindbrain GLP1R circuits for satiety and aversion. Nature 2024;632:585-93.ArticlePubMedPDF
  • 105. Merchenthaler I, Lane M, Shughrue P. Distribution of prepro-glucagon and glucagon-like peptide-1 receptor messenger RNAs in the rat central nervous system. J Comp Neurol 1999;403:261-80.ArticlePubMed
  • 106. Reddy IA, Pino JA, Weikop P, Osses N, Sorensen G, Bering T, et al. Glucagon-like peptide 1 receptor activation regulates cocaine actions and dopamine homeostasis in the lateral septum by decreasing arachidonic acid levels. Transl Psychiatry 2016;6:e809.ArticlePubMedPMCPDF
  • 107. Terrill SJ, Jackson CM, Greene HE, Lilly N, Maske CB, Vallejo S, et al. Role of lateral septum glucagon-like peptide 1 receptors in food intake. Am J Physiol Regul Integr Comp Physiol 2016;311:R124-32.ArticlePubMedPMC
  • 108. Lu Y, Wang L, Luo F, Savani R, Rossi MA, Pang ZP. Dorsolateral septum GLP-1R neurons regulate feeding via lateral hypothalamic projections. Mol Metab 2024;85:101960.ArticlePubMedPMC
  • 109. Chen Z, Deng X, Shi C, Jing H, Tian Y, Zhong J, et al. GLP-1R-positive neurons in the lateral septum mediate the anorectic and weight-lowering effects of liraglutide in mice. J Clin Invest 2024;134:e178239.ArticlePubMedPMC
  • 110. Harasta AE, Power JM, von Jonquieres G, Karl T, Drucker DJ, Housley GD, et al. Septal glucagon-like peptide 1 receptor expression determines suppression of cocaine-induced behavior. Neuropsychopharmacology 2015;40:1969-78.ArticlePubMedPMCPDF
  • 111. Xie Y, Choi T, Al-Aly Z. Mapping the effectiveness and risks of GLP-1 receptor agonists. Nat Med 2025;31:951-62.ArticlePubMedPDF
  • 112. Klausen MK, Thomsen M, Wortwein G, Fink-Jensen A. The role of glucagon-like peptide 1 (GLP-1) in addictive disorders. Br J Pharmacol 2022;179:625-41.ArticlePubMedPDF
  • 113. Klausen MK, Kuzey T, Pedersen JN, Justesen SK, Rasmussen L, Knorr UB, et al. Does semaglutide reduce alcohol intake in Danish patients with alcohol use disorder and comorbid obesity? Trial protocol of a randomised, double-blinded, placebo-controlled clinical trial (the SEMALCO trial). BMJ Open 2025;15:e086454.ArticlePubMedPMC
  • 114. Melson E, Ashraf U, Papamargaritis D, Davies MJ. What is the pipeline for future medications for obesity? Int J Obes (Lond) 2025;49:433-51.ArticlePubMedPDF
  • 115. Petersen J, Ludwig MQ, Juozaityte V, Ranea-Robles P, Svendsen C, Hwang E, et al. GLP-1-directed NMDA receptor antagonism for obesity treatment. Nature 2024;629:1133-41.ArticlePubMedPMCPDF
  • 116. Griffith DA, Edmonds DJ, Fortin JP, Kalgutkar AS, Kuzmiski JB, Loria PM, et al. A small-molecule oral agonist of the human glucagon-like peptide-1 receptor. J Med Chem 2022;65:8208-26.PubMedPMC
  • 117. Zhu X, Fowler MJ, Wells QS, Stafford JM, Gannon M. Predicting responsiveness to GLP-1 pathway drugs using real-world data. BMC Endocr Disord 2024;24:269.ArticlePubMedPMCPDF
  • 118. Tokgoz S, Boss M, Jansen TJ, Meijer R, Frielink C, van Bon AC, et al. Activation of the HPA axis does not explain nonresponsiveness to GLP-1R agonist treatment in individuals with type 2 diabetes. Diabetes 2025;74:212-22.ArticlePubMedPDF
  • 119. Wilding JP, Batterham RL, Davies M, Van Gaal LF, Kandler K, Konakli K, et al. Weight regain and cardiometabolic effects after withdrawal of semaglutide: the STEP 1 trial extension. Diabetes Obes Metab 2022;24:1553-64.PubMedPMC
  • 120. Abdullah Bin Ahmed I. A comprehensive review on weight gain following discontinuation of glucagon-like peptide-1 receptor agonists for obesity. J Obes 2024;2024:8056440.PubMedPMC

Figure & Data

References

    Citations

    Citations to this article as recorded by  

      • PubReader PubReader
      • ePub LinkePub Link
      • Cite this Article
        Cite this Article
        export Copy Download
        Close
        Download Citation
        Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

        Format:
        • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
        • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
        Include:
        • Citation for the content below
        Glucagon-Like Peptide-1 and Hypothalamic Regulation of Satiation: Cognitive and Neural Insights from Human and Animal Studies
        Diabetes Metab J. 2025;49(3):333-347.   Published online May 1, 2025
        Close
      • XML DownloadXML Download
      Figure
      • 0
      • 1
      • 2
      Glucagon-Like Peptide-1 and Hypothalamic Regulation of Satiation: Cognitive and Neural Insights from Human and Animal Studies
      Image Image Image
      Fig. 1. Overview of hypothalamic glucagon-like peptide-1 receptor (GLP-1R) neurons. PVH, paraventricular hypothalamic nucleus; Oxt, oxytocin-expressing; Crh, corticotropin-releasing hormone-expressing; MC4R, melanocortin 4 receptor-expressing; DMH, dorsomedial hypothalamus; LepR, leptin-receptor-expressing; LH, lateral hypothalamus; dDMH, dorsal DMH; vDMH, ventral DMH; GLP-1, glucagon-like peptide-1; ARC, arcuate nucleus; AgRP, agouti-related peptide; POMC, pro-opiomelanocortin; TRH, thyrotropin-releasing hormone-expressing; VMH, ventromedial hypothalamus; i.p., intraperitoneal.
      Fig. 2. Feed-forward cognitive control of satiation. The role of dorsomedial hypothalamus (DMH) glucagon-like peptide-1 receptor (GLP-1R) neurons regulating cognitive satiation can be likened to stepping on a vehicle’s brake in response to traffic signals— a preemptive action taken before reaching the actual stop goal, reflecting a feed-forward control mechanism. These neurons project inhibitory signals to arcuate nucleus (ARC) agouti-related peptide (AgRP) neurons, ultimately contributing to meal termination. Notably, glucagon-like peptide-1 receptor agonists (GLP-1RAs) can potentiate the activity of DMH GLP-1R neurons.
      Graphical abstract
      Glucagon-Like Peptide-1 and Hypothalamic Regulation of Satiation: Cognitive and Neural Insights from Human and Animal Studies
      Brain region Mice GLP-1R transgene [65]a Rat GLP-1R immunocytochemistry [66]a Monkey GLP-1R ISH [67]a Direct injection of GLP-1RA to brain region Disruption of GLP-1R in the area GLP-1RA i.p. injection +, Disruption of GLP-1R in the area Calcium signal of brain region GLP-1R neurons Neuromodulation of brain region GLP-1R neurons
      GLP-1RA i.p. injection effects on brain region GLP-1R neurons
      Activation Inhibition
      DMH ++ +++ ++ (Exendin-4) (DMH glp1r KD or ablation) (i.p. Exendin-4+glp1r KD) Neural activity ↑at pre-ingestive, or ingestive stages Bout duration↓ Bout duration↑ (Exendin-4)
      Blood glucose level ↓ B.W.↑ |Δ Food intake| ↓ Number of bout↓ Food intake↑ Potentiate DMH GLP-1R neurons’ calcium signals
      (Liraglutide) Food intake↑ |Δ B.W. | ↓ Latency to ingestion termination ↓
      B.W. ~ Plasma TG↑ |Δ E.E. |↓
      Food intake ~ E.E. ↓ |Δ RER |↓
      UCP1↓
      Feed pattern disruption
      ARC +++ ++++ ++++ (GLP-1, exendin-4, liraglutide) (POMC glp1r KD) (i.p. Exendin-4+POMC glp1r KD) Food intake ↓ (Liraglutide, semaglutide)
      B.W. ↓ B.W. ~ |Δ Food intake| ~ Increase excitatory tone to POMC neurons
      Food intake↓ HFD-induced weight gain ↑ (i.p. Liraglutide+ARC glp1r antagonist)
      Food intake ~ |Δ Food intake| ↓
      E.E. ~
      PVH +++ +++ +++ (GLP-1, exendin-4, liraglutide) (Sim1 glp1r KD) (i.p. Exendin-4+Sim1 glp1r KD) Neural activity ↑at pre-ingestive, or ingestive stages Food intake ↓ Food intake ↑
      B.W. ↓ B.W. ~ |Δ Food intake| ~
      Food intake↓ HFD-induced weight gain ~ (i.p. Liraglutide+PVH glp1r antagonist)
      Food intake ~ |Δ Food intake| ~
      E.E. ↓
      (Juvenile PVH glp1r KO)
      B.W.↑
      Food intake↑
      E.E. ~
      (PVH glp1r antagonist)
      B.W.↑
      LH ++ ++ ++ (Exendin-4) NAc GLP-1R expression ↑
      Lever-pressing ↓
      (Liraglutide)
      B.W. ↓
      Food intake↓
      VMH +++ - - (Exendin-4) (VMH glp1r KD) (Exendin-4) - - - -
      B.W. ↓ Food intake ~ |Δ Food intake| ~
      Food intake↓ B.W. ~ (Liraglutide)
      E.E. ↓ E.E. ~ |Δ B.W. | ~
      (Liraglutide)
      B.W. ↓
      Food intake ~
      Table 1. Animal studies of GLP-1R, GLP-1R neurons, GLP-1RA effects on hypothalamus

      GLP-1R, glucagon-like peptide-1 receptor; GLP-1RA, glucagon-like peptide-1 receptor agonist; ISH, in situ hybridization; DMH, dorsomedial hypothalamus; B.W., body weight; KD, knockdown; TG, triglyceride; E.E., energy expenditure; UCP1, uncoupling protein 1; i.p., intraperitoneal; RER, resting energy requirement; ARC, arcuate nucleus; POMC, pro-opiomelanocortin; HFD, high-fat diet; PVH, paraventricular hypothalamic nucleus; KO, knockout; Sim1, single minded-1; LH, lateral hypothalamus; NAc, nucleus accumbens; VMH, ventromedial hypothalamus.

      GLP-1R expression detected by transgene expression in mice [65], density of GLP-1R-immunoreactive fibers in each brain region determined by rat immunocytochemistry [66], GLP-1R expression determined by ISH in non-human primates [67].

      Park JS, Kim KS, Choi HJ. Glucagon-Like Peptide-1 and Hypothalamic Regulation of Satiation: Cognitive and Neural Insights from Human and Animal Studies. Diabetes Metab J. 2025;49(3):333-347.
      Received: Feb 09, 2025; Accepted: Apr 16, 2025
      DOI: https://doi.org/10.4093/dmj.2025.0106.

      Diabetes Metab J : Diabetes & Metabolism Journal
      Close layer
      TOP