The glucagon-like peptide-1 (GLP-1) analogue semaglutide reduces alcohol drinking and modulates central GABA neurotransmission

Vicky Chuong, Mehdi Farokhnia, Sophia Khom, Claire L Pince, Sophie K Elvig, Roman Vlkolinsky, Renata Cn Marchette, George F Koob, Marisa Roberto, Leandro F Vendruscolo, Lorenzo Leggio, Vicky Chuong, Mehdi Farokhnia, Sophia Khom, Claire L Pince, Sophie K Elvig, Roman Vlkolinsky, Renata Cn Marchette, George F Koob, Marisa Roberto, Leandro F Vendruscolo, Lorenzo Leggio

Abstract

Growing evidence indicates that the glucagon-like peptide-1 (GLP-1) system is involved in the neurobiology of addictive behaviors, and GLP-1 analogues may be used for the treatment of alcohol use disorder (AUD). Here, we examined the effects of semaglutide, a long-acting GLP-1 analogue, on biobehavioral correlates of alcohol use in rodents. A drinking-in-the-dark procedure was used to test the effects of semaglutide on binge-like drinking in male and female mice. We also tested the effects of semaglutide on binge-like and dependence-induced alcohol drinking in male and female rats, as well as acute effects of semaglutide on spontaneous inhibitory postsynaptic currents (sIPSCs) from central amygdala (CeA) and infralimbic cortex (ILC) neurons. Semaglutide dose-dependently reduced binge-like alcohol drinking in mice; a similar effect was observed on the intake of other caloric/noncaloric solutions. Semaglutide also reduced binge-like and dependence-induced alcohol drinking in rats. Semaglutide increased sIPSC frequency in CeA and ILC neurons from alcohol-naive rats, suggesting enhanced GABA release, but had no overall effect on GABA transmission in alcohol-dependent rats. In conclusion, the GLP-1 analogue semaglutide decreased alcohol intake across different drinking models and species and modulated central GABA neurotransmission, providing support for clinical testing of semaglutide as a potentially novel pharmacotherapy for AUD.

Keywords: Addiction; Endocrinology; Neuroscience.

Figures

Figure 1. Semaglutide reduces binge-like alcohol drinking…
Figure 1. Semaglutide reduces binge-like alcohol drinking in mice.
(A) Semaglutide reduced alcohol intake (g/kg of body weight) in mice drinking sweet alcohol. Males (n = 8); females (n = 7). (B) Semaglutide reduced alcohol intake (g/kg of body weight) in mice drinking unsweet alcohol; female mice drank significantly more alcohol than males. Males (n = 8); females (n = 8). (C) Semaglutide reduced fluid intake (mL/kg of body weight) in mice drinking a sweet solution not containing alcohol. Males (n = 8); females (n = 6). Separate cohorts of mice were used to test the effects of semaglutide on the consumption of each drinking solution. Data are expressed as mean ± SEM and were analyzed using 2-way repeated-measures ANOVA. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 versus vehicle. Individual values are presented for males (♂) and females (♀).
Figure 2. Semaglutide reduces drinking of noncaloric…
Figure 2. Semaglutide reduces drinking of noncaloric and caloric solutions not containing alcohol in mice.
(A and B) Semaglutide reduced fluid intake (mL/kg of body weight) in mice drinking water or a saccharin-sweetened noncaloric solution. (C and D) Semaglutide reduced calorie intake (Kcal/kg of body weight) in mice drinking an unsweet carbohydrate (maltodextrin) solution or an unsweet fat (corn oil) emulsion. Separate cohorts of mice were used to test the effects of semaglutide on the consumption of each drinking solution (n = 8, 4 per sex, per condition). Data are expressed as mean ± SEM and were analyzed using 1-way repeated-measures ANOVA. **P < 0.01, ***P < 0.001, ****P < 0.0001 versus vehicle. Individual values are presented for males (♂) and females (♀).
Figure 3. Semaglutide reduces operant alcohol self-administration…
Figure 3. Semaglutide reduces operant alcohol self-administration in rats.
(A) Semaglutide dose-dependently reduced sweet alcohol self-administration (binge-like drinking) in rats. (B) Semaglutide did not reduce water self-administration in nondependent rats (significant Dose effect, but no significant post hoc differences); female nondependent rats self-administered significantly more water than males. Nondependent males (n = 10); nondependent females (n = 10). (C) Semaglutide only at the highest dose (0.1 mg/kg) reduced unsweet alcohol self-administration (dependence-induced drinking) in rats. (D) Semaglutide had no effect on water self-administration in alcohol-dependent rats; male dependent rats self-administered significantly more water than females. Dependent males (n = 11); dependent females (n = 11). Data are expressed as mean ± SEM and were analyzed using 2-way repeated-measures ANOVA. **P < 0.01, ****P < 0.0001 versus vehicle. Individual values are presented for males (♂) and females (♀).
Figure 4. Semaglutide increased GABA transmission in…
Figure 4. Semaglutide increased GABA transmission in central nucleus of the amygdala (CeA) neurons from alcohol-naive rats but had mixed effects in alcohol-dependent rats.
(A) Representative spontaneous inhibitory postsynaptic current (sIPSC) traces during baseline control (upper panel) conditions and during superfusion of 100 nM semaglutide (lower panel). (BE) Bar charts summarize the effects of semaglutide (100nM) on sIPSC frequencies (B), amplitudes (C), rise times (D), and decay times (E) from 10 to 15 neurons from alcohol-naive (gray bars) and alcohol-dependent rats (red bars). Data are expressed as mean ± SEM. Differences between semaglutide and baseline control conditions (dashed lines) were analyzed using 1-sample Student’s t tests (**P < 0.01). Differences of semaglutide effects on selected parameters between alcohol-naive and alcohol-dependent rats were analyzed using unpaired Student’s t tests ($P < 0.05). Data were generated from 6 alcohol-naive and 8 alcohol-dependent rats, from 2 separate chronic, intermittent, alcohol vapor exposure cohorts.
Figure 5. Semaglutide increased GABA transmission in…
Figure 5. Semaglutide increased GABA transmission in pyramidal neurons in layer 5 of the infralimbic cortex (ILC) from alcohol-naive rats but had mixed effects in alcohol-dependent rats.
(A) Representative spontaneous inhibitory postsynaptic currents (sIPSC) traces during baseline control (upper panel) conditions and during superfusion of 100 nM semaglutide (lower panel). (BE) Bar charts summarize the effects of semaglutide (100nM) on sIPSC frequencies (B), amplitudes (C), rise times (D), and decay times (E) from 9 to 12 neurons from alcohol-naive (gray bars) and alcohol-dependent rats (red bars). Data are expressed as mean ± SEM. Differences between semaglutide and baseline control conditions (dashed lines) were analyzed using 1-sample Student’s t tests (**P < 0.01). Differences of semaglutide effects on selected parameters between alcohol-naive and alcohol-dependent rats were calculated using unpaired Student’s t tests ($P < 0.05). Data were generated from 5 alcohol-naive and 7 alcohol-dependent rats, from 2 separate chronic, intermittent, alcohol vapor exposure cohorts.

References

    1. Witkiewitz K, et al. Advances in the science and treatment of alcohol use disorder. Sci Adv. 2019;5(9):eaax4043. doi: 10.1126/sciadv.aax4043.
    1. Moore CF, et al. Pathological overeating: emerging evidence for a compulsivity construct. Neuropsychopharmacology. 2017;42(7):1375–1389. doi: 10.1038/npp.2016.269.
    1. Volkow ND, et al. The addictive dimensionality of obesity. Biol Psychiatry. 2013;73(9):811–818. doi: 10.1016/j.biopsych.2012.12.020.
    1. Farokhnia M, et al. Ghrelin: From a gut hormone to a potential therapeutic target for alcohol use disorder. Physiol Behav. 2019;204:49–57. doi: 10.1016/j.physbeh.2019.02.008.
    1. Jerlhag E. Gut-brain axis and addictive disorders: A review with focus on alcohol and drugs of abuse. Pharmacol Ther. 2019;196:1–14. doi: 10.1016/j.pharmthera.2018.11.005.
    1. Klausen MK, et al. The role of glucagon-like peptide 1 (GLP-1) in addictive disorders. Br J Pharmacol. 2022;179(4):625–641. doi: 10.1111/bph.15677.
    1. Novak U, et al. Identical mRNA for preproglucagon in pancreas and gut. Eur J Biochem. 1987;164(3):553–558. doi: 10.1111/j.1432-1033.1987.tb11162.x.
    1. Larsen PJ, et al. Distribution of glucagon-like peptide-1 and other preproglucagon-derived peptides in the rat hypothalamus and brainstem. Neuroscience. 1997;77(1):257–270. doi: 10.1016/S0306-4522(96)00434-4.
    1. Jensen CB, et al. Characterization of the glucagonlike peptide-1 receptor in male mouse brain using a novel antibody and in situ hybridization. Endocrinology. 2018;159(2):665–675. doi: 10.1210/en.2017-00812.
    1. Krieger JP, et al. Vagal mediation of GLP-1’s effects on food intake and glycemia. Physiol Behav. 2015;152(pt b):372–380. doi: 10.1016/j.physbeh.2015.06.001.
    1. Madsbad S. The role of glucagon-like peptide-1 impairment in obesity and potential therapeutic implications. Diabetes Obes Metab. 2014;16(1):9–21. doi: 10.1111/dom.12119.
    1. During MJ, et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat Med. 2003;9(9):1173–1179. doi: 10.1038/nm919.
    1. Kinzig KP, et al. CNS glucagon-like peptide-1 receptors mediate endocrine and anxiety responses to interoceptive and psychogenic stressors. J Neurosci. 2003;23(15):6163–6170. doi: 10.1523/JNEUROSCI.23-15-06163.2003.
    1. Eren-Yazicioglu CY, et al. Can GLP-1 be a target for reward system related disorders? A qualitative synthesis and systematic review analysis of studies on palatable food, drugs of abuse, and alcohol. Front Behav Neurosci. 2021;14:614884. doi: 10.3389/fnbeh.2020.614884.
    1. Guerrero-Hreins E, et al. The therapeutic potential of GLP-1 analogues for stress-related eating and role of GLP-1 in stress, emotion and mood: a review. Prog Neuropsychopharmacol Biol Psychiatry. 2021;110:110303. doi: 10.1016/j.pnpbp.2021.110303.
    1. Trapp S, Brierley DI. Brain GLP-1 and the regulation of food intake: GLP-1 action in the brain and its implications for GLP-1 receptor agonists in obesity treatment. Br J Pharmacol. 2022;179(4):557–570. doi: 10.1111/bph.15638.
    1. Jerlhag E. Alcohol-mediated behaviours and the gut-brain axis; with focus on glucagon-like peptide-1. Brain Res. 2020;1727:146562. doi: 10.1016/j.brainres.2019.146562.
    1. Heppner KM, 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(1):255–267. doi: 10.1210/en.2014-1675.
    1. Pyke C, et al. GLP-1 receptor localization in monkey and human tissue: novel distribution revealed with extensively validated monoclonal antibody. Endocrinology. 2014;155(4):1280–1290. doi: 10.1210/en.2013-1934.
    1. Cork SC, et al. Distribution and characterisation of Glucagon-like peptide-1 receptor expressing cells in the mouse brain. Mol Metab. 2015;4(10):718–731. doi: 10.1016/j.molmet.2015.07.008.
    1. Merchenthaler I, et al. Distribution of pre-pro-glucagon and glucagon-like peptide-1 receptor messenger RNAs in the rat central nervous system. J Comp Neurol. 1999;403(2):261–280. doi: 10.1002/(SICI)1096-9861(19990111)403:2<261::AID-CNE8>;2-5.
    1. Göke R, et al. Distribution of GLP-1 binding sites in the rat brain: evidence that exendin-4 is a ligand of brain GLP-1 binding sites. Eur J Neurosci. 1995;7(11):2294–2300. doi: 10.1111/j.1460-9568.1995.tb00650.x.
    1. Anderberg RH, et al. GLP-1 is both anxiogenic and antidepressant; divergent effects of acute and chronic GLP-1 on emotionality. Psychoneuroendocrinology. 2016;65:54–66. doi: 10.1016/j.psyneuen.2015.11.021.
    1. Hayes MR, et al. Role of the glucagon-like-peptide-1 receptor in the control of energy balance. Physiol Behav. 2010;100(5):503–510. doi: 10.1016/j.physbeh.2010.02.029.
    1. Kenny PJ. Common cellular and molecular mechanisms in obesity and drug addiction. Nat Rev Neurosci. 2011;12(11):638–651. doi: 10.1038/nrn3105.
    1. Dickson SL, et al. The glucagon-like peptide 1 (GLP-1) analogue, exendin-4, decreases the rewarding value of food: a new role for mesolimbic GLP-1 receptors. J Neurosci. 2012;32(14):4812–4820. doi: 10.1523/JNEUROSCI.6326-11.2012.
    1. Ghosal S, et al. Disruption of glucagon-like peptide 1 signaling in sim1 neurons reduces physiological and behavioral reactivity to acute and chronic stress. J Neurosci. 2017;37(1):184–193.
    1. Roberto M, et al. The role of the central amygdala in alcohol dependence. Cold Spring Harb Perspect Med. 2021;11(2):a039339. doi: 10.1101/cshperspect.a039339.
    1. Pfarr S, et al. Losing control: excessive alcohol seeking after selective inactivation of cue-responsive neurons in the infralimbic cortex. J Neurosci. 2015;35(30):10750–10761. doi: 10.1523/JNEUROSCI.0684-15.2015.
    1. Koob GF. Drug addiction: hyperkatifeia/negative reinforcement as a framework for medications development. Pharmacol Rev. 2021;73(1):163–201. doi: 10.1124/pharmrev.120.000083.
    1. Khom S, et al. Alcohol dependence and withdrawal impair serotonergic regulation of GABA transmission in the rat central nucleus of the amygdala. J Neurosci. 2020;40(36):6842–6853. doi: 10.1523/JNEUROSCI.0733-20.2020.
    1. Khom S, et al. Alcohol dependence potentiates substance P/neurokinin-1 receptor signaling in the rat central nucleus of amygdala. Sci Adv. 2020;6(12):eaaz1050. doi: 10.1126/sciadv.aaz1050.
    1. Roberto M, et al. Increased GABA release in the central amygdala of ethanol-dependent rats. J Neurosci. 2004;24(45):10159–10166. doi: 10.1523/JNEUROSCI.3004-04.2004.
    1. Laque A G LDN, et al. Anti-relapse neurons in the infralimbic cortex of rats drive relapse-suppression by drug omission cues. Nat Commun. 2019;10(1):3934. doi: 10.1038/s41467-019-11799-1.
    1. Flores-Ramirez FJ, et al. Blockade of orexin receptors in the infralimbic cortex prevents stress-induced reinstatement of alcohol-seeking behaviour in alcohol-dependent rats. Br J Pharmacol. 2023;180(11):1500–1515. doi: 10.1111/bph.16015.
    1. Patel RR, et al. Ethanol withdrawal-induced adaptations in prefrontal corticotropin releasing factor receptor 1-expressing neurons regulate anxiety and conditioned rewarding effects of ethanol. Mol Psychiatry. 2022;27(8):3441–3451. doi: 10.1038/s41380-022-01642-3.
    1. Varodayan FP, et al. Chronic ethanol induces a pro-inflammatory switch in interleukin-1β regulation of GABAergic signaling in the medial prefrontal cortex of male mice. Brain Behav Immun. 2023;110:125–139. doi: 10.1016/j.bbi.2023.02.020.
    1. Korol SV, et al. GLP-1 and exendin-4 transiently enhance GABAA receptor-mediated synaptic and tonic currents in rat hippocampal CA3 pyramidal neurons. Diabetes. 2015;64(1):79–89. doi: 10.2337/db14-0668.
    1. Rebosio C, et al. Presynaptic GLP-1 receptors enhance the depolarization-evoked release of glutamate and GABA in the mouse cortex and hippocampus. Biofactors. 2018;44(2):148–157. doi: 10.1002/biof.1406.
    1. Fortin SM, 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(533):eaay8071. doi: 10.1126/scitranslmed.aay8071.
    1. Kieffer TJ, et al. Degradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IV. Endocrinology. 1995;136(8):3585–3596. doi: 10.1210/endo.136.8.7628397.
    1. Donnelly D. The structure and function of the glucagon-like peptide-1 receptor and its ligands. Br J Pharmacol. 2012;166(1):27–41. doi: 10.1111/j.1476-5381.2011.01687.x.
    1. Knudsen LB, Lau J. The discovery and development of liraglutide and semaglutide. Front Endocrinol (lausanne) 2019;10:155. doi: 10.3389/fendo.2019.00155.
    1. Marty VN, et al. Long-acting glucagon-like peptide-1 receptor agonists suppress voluntary alcohol intake in male wistar rats. Front Neurosci. 2020;14(1378):599646:599646.
    1. O’Neil PM, et al. Efficacy and safety of semaglutide compared with liraglutide and placebo for weight loss in patients with obesity: a randomised, double-blind, placebo and active controlled, dose-ranging, phase 2 trial. Lancet. 2018;392(10148):637–649. doi: 10.1016/S0140-6736(18)31773-2.
    1. Lingvay I, et al. Superior weight loss with once-weekly semaglutide versus other glucagon-like peptide-1 receptor agonists is independent of gastrointestinal adverse events. BMJ Open Diabetes Res Care. 2020;8(2):e001706. doi: 10.1136/bmjdrc-2020-001706.
    1. Wilding JPH, et al. Once-weekly semaglutide in adults with overweight or obesity. N Engl J Med. 2021;384(11):989. doi: 10.1056/NEJMoa2032183.
    1. Lau J, et al. Discovery of the once-weekly glucagon-like peptide-1 (GLP-1) analogue semaglutide. J Med Chem. 2015;58(18):7370–7380. doi: 10.1021/acs.jmedchem.5b00726.
    1. Jensen L, et al. Absorption, metabolism and excretion of the GLP-1 analogue semaglutide in humans and nonclinical species. Eur J Pharm Sci. 2017;104:31–41. doi: 10.1016/j.ejps.2017.03.020.
    1. Christou GA, et al. Semaglutide as a promising antiobesity drug. Obes Rev. 2019;20(6):805–815. doi: 10.1111/obr.12839.
    1. Bucheit JD, et al. Oral semaglutide: a review of the first oral glucagon-like peptide 1 receptor agonist. Diabetes Technol Ther. 2020;22(1):10–18. doi: 10.1089/dia.2019.0185.
    1. Molina PE, Nelson S. Binge drinking’s effects on the body. Alcohol Res. 2018;39(1):99–109.
    1. Pérez-García JM, et al. Effects of binge drinking during adolescence and emerging adulthood on the brain: A systematic review of neuroimaging studies. Neurosci Biobehav Rev. 2022;137:104637. doi: 10.1016/j.neubiorev.2022.104637.
    1. Vendruscolo LF, Roberts AJ. Operant alcohol self-administration in dependent rats: focus on the vapor model. Alcohol. 2014;48(3):277–286. doi: 10.1016/j.alcohol.2013.08.006.
    1. Volcko KL, et al. Control of water intake by a pathway from the nucleus of the solitary tract to the paraventricular hypothalamic nucleus. Appetite. 2022;172:105943. doi: 10.1016/j.appet.2022.105943.
    1. McKay NJ, et al. Endogenous glucagon-like peptide-1 reduces drinking behavior and is differentially engaged by water and food intakes in rats. J Neurosci. 2014;34(49):16417–16423. doi: 10.1523/JNEUROSCI.3267-14.2014.
    1. McKay NJ, Daniels D. Glucagon-like peptide-1 receptor agonist administration suppresses both water and saline intake in rats. J Neuroendocrinol. 2013;25(10):929–938. doi: 10.1111/jne.12086.
    1. Gutzwiller JP, et al. Glucagon-like peptide-1 is involved in sodium and water homeostasis in humans. Digestion. 2006;73(2-3):142–150. doi: 10.1159/000094334.
    1. Winzeler B, et al. Effects of glucagon-like peptide-1 receptor agonists on fluid intake in healthy volunteers. Endocrine. 2020;70(2):292–298. doi: 10.1007/s12020-020-02394-2.
    1. Winzeler BF, et al. GLP1 receptor agonists reduce fluid intake in primary polydipsia. J Endocr Soc. 2021;5(suppl_1):A514–A515. doi: 10.1210/jendso/bvab048.1052.
    1. Takai S, et al. Glucagon-like peptide-1 is specifically involved in sweet taste transmission. FASEB J. 2015;29(6):2268–2280. doi: 10.1096/fj.14-265355.
    1. Martin B, et al. Modulation of taste sensitivity by GLP-1 signaling in taste buds. Ann N Y Acad Sci. 2009;1170:98–101. doi: 10.1111/j.1749-6632.2009.03920.x.
    1. Kreuch D, et al. Gut mechanisms linking intestinal sweet sensing to glycemic control. Front Endocrinol (lausanne) 2018;9:741:741.
    1. Ohtsu Y, et al. Diverse signaling systems activated by the sweet taste receptor in human GLP-1-secreting cells. Mol Cell Endocrinol. 2014;394(1-2):70–79. doi: 10.1016/j.mce.2014.07.004.
    1. Barrera JG, et al. GLP-1 and energy balance: an integrated model of short-term and long-term control. Nat Rev Endocrinol. 2011;7(9):507–516. doi: 10.1038/nrendo.2011.77.
    1. Velásquez DA, et al. Central GLP-1 actions on energy metabolism. Vitam Horm. 2010;84:303–317. doi: 10.1016/B978-0-12-381517-0.00011-4.
    1. Krieger JP, et al. Novel role of GLP-1 receptor signaling in energy expenditure during chronic high fat diet feeding in rats. Physiol Behav. 2018;192:194–199. doi: 10.1016/j.physbeh.2018.03.037.
    1. Näslund E, et al. Energy intake and appetite are suppressed by glucagon-like peptide-1 (GLP-1) in obese men. Int J Obes Relat Metab Disord. 1999;23(3):304–311. doi: 10.1038/sj.ijo.0800818.
    1. Bergmann NC, et al. Semaglutide for the treatment of overweight and obesity: A review. Diabetes Obes Metab. 2023;25(1):18–35. doi: 10.1111/dom.14863.
    1. Leggio L, et al. Role of feeding-related pathways in alcohol dependence: A focus on sweet preference, NPY, and ghrelin. Alcohol Clin Exp Res. 2011;35(2):194–202. doi: 10.1111/j.1530-0277.2010.01334.x.
    1. Cameron F, et al. Phentermine and topiramate extended release (Qsymia™): first global approval. Drugs. 2012;72(15):2033–2042. doi: 10.2165/11640860-000000000-00000.
    1. Reus VI, et al. The American Psychiatric Association practice guideline for the pharmacological treatment of patients with alcohol use disorder. Am J Psychiatry. 2018;175(1):86–90. doi: 10.1176/appi.ajp.2017.1750101.
    1. Perry C, et al. The management of substance use disorders: synopsis of the 2021 U.S. Department of Veterans Affairs and U.S. Department of Defense Clinical Practice Guideline. Ann Intern Med. 2022;175(5):720–731. doi: 10.7326/M21-4011.
    1. Serafine KM, et al. Converging vulnerability factors for compulsive food and drug use. Neuropharmacology. 2021;196:108556. doi: 10.1016/j.neuropharm.2021.108556.
    1. Traversy G, Chaput JP. Alcohol consumption and obesity: an update. Curr Obes Rep. 2015;4(1):122–130. doi: 10.1007/s13679-014-0129-4.
    1. Mahli A, Hellerbrand C. Alcohol and obesity: a dangerous association for fatty liver disease. Dig Dis. 2016;34 Suppl 1:32–39. doi: 10.1159/000447279.
    1. Piano MR. Alcohol’s effects on the cardiovascular system. Alcohol Res. 2017;38(2):219–241.
    1. Thiele TE, et al. Central infusion of GLP-1, but not leptin, produces conditioned taste aversions in rats. Am J Physiol. 1997;272(2):R726–R730.
    1. Kinzig KP, et al. The diverse roles of specific GLP-1 receptors in the control of food intake and the response to visceral illness. J Neurosci. 2002;22(23):10470–10476. doi: 10.1523/JNEUROSCI.22-23-10470.2002.
    1. Costa A, et al. Anorectic and aversive effects of GLP-1 receptor agonism are mediated by brainstem cholecystokinin neurons, and modulated by GIP receptor activation. Mol Metab. 2022;55:101407. doi: 10.1016/j.molmet.2021.101407.
    1. Kanoski SE, et al. The role of nausea in food intake and body weight suppression by peripheral GLP-1 receptor agonists, exendin-4 and liraglutide. Neuropharmacology. 2012;62(5):1916–1927.
    1. Scott KA, Moran TH. The GLP-1 agonist exendin-4 reduces food intake in nonhuman primates through changes in meal size. Am J Physiol Regul Integr Comp Physiol. 2007;293(3):R983–R987. doi: 10.1152/ajpregu.00323.2007.
    1. Thomsen M, et al. Effects of glucagon-like peptide 1 analogs on alcohol intake in alcohol-preferring vervet monkeys. Psychopharmacology. 2019;236(2):603–611. doi: 10.1007/s00213-018-5089-z.
    1. Ghidewon M, et al. Growth differentiation factor 15 (GDF15) and semaglutide inhibit food intake and body weight through largely distinct, additive mechanisms. Diabetes Obes Metab. 2022;24(6):1010–1020. doi: 10.1111/dom.14663.
    1. Dossat AM, et al. Glucagon-like peptide 1 receptors in nucleus accumbens affect food intake. J Neurosci. 2011;31(41):14453–14457. doi: 10.1523/JNEUROSCI.3262-11.2011.
    1. Alhadeff AL, et al. GLP-1 neurons in the nucleus of the solitary tract project directly to the ventral tegmental area and nucleus accumbens to control for food intake. Endocrinology. 2012;153(2):647–658. doi: 10.1210/en.2011-1443.
    1. Richard JE, et al. Activation of the GLP-1 receptors in the nucleus of the solitary tract reduces food reward behavior and targets the mesolimbic system. PLoS One. 2015;10(3):e0119034. doi: 10.1371/journal.pone.0119034.
    1. Hayes MR, Schmidt HD. GLP-1 influences food and drug reward. Curr Opin Behav Sci. 2016;9:66–70. doi: 10.1016/j.cobeha.2016.02.005.
    1. Egecioglu E, et al. The glucagon-like peptide 1 analogue Exendin-4 attenuates alcohol mediated behaviors in rodents. Psychoneuroendocrinology. 2013;38(8):1259–1270. doi: 10.1016/j.psyneuen.2012.11.009.
    1. Shirazi RH, et al. Gut peptide GLP-1 and its analogue, Exendin-4, decrease alcohol intake and reward. PLoS One. 2013;8(4):e61965. doi: 10.1371/journal.pone.0061965.
    1. Schmidt HD, et al. Glucagon-like peptide-1 receptor activation in the ventral tegmental area decreases the reinforcing efficacy of cocaine. Neuropsychopharmacology. 2016;41(7):1917–1928. doi: 10.1038/npp.2015.362.
    1. Wang X-F, et al. Endogenous glucagon-like peptide-1 suppresses high-fat food intake by reducing synaptic drive onto mesolimbic dopamine neurons. Cell Rep. 2015;12(5):726–733. doi: 10.1016/j.celrep.2015.06.062.
    1. Fortin SM, Roitman MF. Central GLP-1 receptor activation modulates cocaine-evoked phasic dopamine signaling in the nucleus accumbens core. Physiol Behav. 2017;176:17–25. doi: 10.1016/j.physbeh.2017.03.019.
    1. Hernandez NS, et al. Glucagon-like peptide-1 receptor activation in the ventral tegmental area attenuates cocaine seeking in rats. Neuropsychopharmacology. 2018;43(10):2000–2008. doi: 10.1038/s41386-018-0010-3.
    1. Vallöf D, et al. Brain region specific glucagon-like peptide-1 receptors regulate alcohol-induced behaviors in rodents. Psychoneuroendocrinology. 2019;103:284–295. doi: 10.1016/j.psyneuen.2019.02.006.
    1. Colvin KJ, et al. Brain site-specific inhibitory effects of the GLP-1 analogue exendin-4 on alcohol intake and operant responding for palatable food. Int J Mol Sci. 2020;21(24):9710. doi: 10.3390/ijms21249710.
    1. Dixon TN, et al. Glucagon-like peptide-1 receptor signaling in the ventral tegmental area reduces alcohol self-administration in male rats. Alcohol Clin Exp Res. 2020;44(10):2118–2129. doi: 10.1111/acer.14437.
    1. Wharton S, et al. Gastrointestinal tolerability of once-weekly semaglutide 2.4 mg in adults with overweight or obesity, and the relationship between gastrointestinal adverse events and weight loss. Diabetes, Obesity and Metabolism. 2022;24(1):94–105. doi: 10.1111/dom.14551.
    1. Falk DE, et al. Evaluation of drinking risk levels as outcomes in alcohol pharmacotherapy trials: a secondary analysis of 3 randomized clinical trials. JAMA Psychiatry. 2019;76(4):374–381. doi: 10.1001/jamapsychiatry.2018.3079.
    1. Hasin DS, et al. Change in non-abstinent WHO drinking risk levels and alcohol dependence: a 3 year follow-up study in the US general population. Lancet Psychiatry. 2017;4(6):469–476. doi: 10.1016/S2215-0366(17)30130-X.
    1. Khom S, et al. Alcohol dependence and withdrawal increase sensitivity of central amygdalar GABAergic synapses to the glucocorticoid receptor antagonist mifepristone in male rats. Neurobiol Dis. 2022;164:105610. doi: 10.1016/j.nbd.2022.105610.
    1. Vendruscolo LF, et al. Glucocorticoid receptor antagonism decreases alcohol seeking in alcohol-dependent individuals. J Clin Invest. 2015;125(8):3193–3197. doi: 10.1172/JCI79828.
    1. He Z, 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;28:120–134. doi: 10.1016/j.molmet.2019.07.008.
    1. Babateen O, et al. Liraglutide modulates GABAergic signaling in rat hippocampal CA3 pyramidal neurons predominantly by presynaptic mechanism. BMC Pharmacol Toxicol. 2017;18(1):83. doi: 10.1186/s40360-017-0191-0.
    1. Korol SV, et al. The GLP-1 receptor agonist exendin-4 and diazepam differentially regulate GABAA receptor-mediated tonic currents in rat hippocampal CA3 pyramidal neurons. PLoS One. 2015;10(4):e0124765. doi: 10.1371/journal.pone.0124765.
    1. Doyle ME, Egan JM. Mechanisms of action of glucagon-like peptide 1 in the pancreas. Pharmacol Ther. 2007;113(3):546–593. doi: 10.1016/j.pharmthera.2006.11.007.
    1. Lønsmann I, Bak LK. Potential role of adenylyl cyclase 8 signaling complexes in regulating insulin secretion from pancreatic beta cells. Cell Signal. 2020;72:109635. doi: 10.1016/j.cellsig.2020.109635.
    1. Augier E, et al. A molecular mechanism for choosing alcohol over an alternative reward. Science. 2018;360(6395):1321–1326. doi: 10.1126/science.aao1157.
    1. Jimenez VA, et al. Synaptic adaptations in the central amygdala and hypothalamic paraventricular nucleus associated with protracted ethanol abstinence in male rhesus monkeys. Neuropsychopharmacology. 2019;44(5):982–993. doi: 10.1038/s41386-018-0290-7.
    1. Bajo M, et al. Innate immune factors modulate ethanol interaction with GABAergic transmission in mouse central amygdala. Brain Behav Immun. 2014;40:191–202. doi: 10.1016/j.bbi.2014.03.007.
    1. Roberto M, et al. Cellular and behavioral interactions of gabapentin with alcohol dependence. J Neurosci. 2008;28(22):5762–5771. doi: 10.1523/JNEUROSCI.0575-08.2008.
    1. Leggio L, et al. Typologies of alcohol dependence. From Jellinek to genetics and beyond. Neuropsychol Rev. 2009;19(1):115–129. doi: 10.1007/s11065-008-9080-z.
    1. Klausen MK, et al. Exenatide once weekly for alcohol use disorder investigated in a randomized, placebo-controlled clinical trial. JCI Insight. 2022;7(19):e159863. doi: 10.1172/jci.insight.159863.
    1. Wium-Andersen IK, et al. Use of GLP-1 receptor agonists and subsequent risk of alcohol-related events. A nationwide register-based cohort and self-controlled case series study. Basic Clin Pharmacol Toxicol. 2022;131(5):372–379. doi: 10.1111/bcpt.13776.
    1. Farokhnia M, et al. The glucagon-like peptide-1 system is modulated by acute and chronic alcohol exposure: Findings from human laboratory experiments and a post-mortem brain study. Addict Biol. 2022;27(5):e13211.
    1. Suchankova P, et al. The glucagon-like peptide-1 receptor as a potential treatment target in alcohol use disorder: evidence from human genetic association studies and a mouse model of alcohol dependence. Transl Psychiatry. 2015;5(6):e583. doi: 10.1038/tp.2015.68.
    1. Farokhnia M, et al. Differential association between the GLP1R gene variants and brain functional connectivity according to the severity of alcohol use. Sci Rep. 2022;12(1):13027. doi: 10.1038/s41598-022-17190-3.
    1. O’Sullivan SJ, et al. Single cell scale neuronal and glial gene expression and putative cell phenotypes and networks in the nucleus tractus solitarius in an alcohol withdrawal time series. Front Syst Neurosci. 2021;15:739790. doi: 10.3389/fnsys.2021.739790.
    1. Rhodes JS, et al. Evaluation of a simple model of ethanol drinking to intoxication in C57BL/6J mice. Physiol Behav. 2005;84(1):53–63. doi: 10.1016/j.physbeh.2004.10.007.
    1. Farokhnia M, et al. Spironolactone as a potential new pharmacotherapy for alcohol use disorder: convergent evidence from rodent and human studies. Mol Psychiatry. 2022;27(11):4642–4652. doi: 10.1038/s41380-022-01736-y.
    1. Hefner K, Holmes A. An investigation of the behavioral actions of ethanol across adolescence in mice. Psychopharmacology (Berl) 2007;191(2):311–322. doi: 10.1007/s00213-006-0646-2.
    1. Ji D, et al. Effects of naltrexone, duloxetine, and a corticotropin-releasing factor type 1 receptor antagonist on binge-like alcohol drinking in rats. Behav Pharmacol. 2008;19(1):1–12. doi: 10.1097/FBP.0b013e3282f3cf70.
    1. Zallar LJ, et al. Ghrelin receptor deletion reduces binge-like alcohol drinking in rats. J Neuroendocrinol. 2019;31(7):e12663.
    1. McGinn MA, et al. Glucocorticoid receptor modulators decrease alcohol self-administration in male rats. Neuropharmacology. 2021;188:108510. doi: 10.1016/j.neuropharm.2021.108510.
    1. Chieng BC, et al. Characterization of neurons in the rat central nucleus of the amygdala: cellular physiology, morphology, and opioid sensitivity. J Comp Neurol. 2006;497(6):910–927. doi: 10.1002/cne.21025.
    1. Gabery S, et al. Semaglutide lowers body weight in rodents via distributed neural pathways. JCI Insight. 2020;5(6):e133429. doi: 10.1172/jci.insight.133429.

Source: PubMed

3
Abonner