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

P Suchankova, J Yan, M L Schwandt, B L Stangl, E C Caparelli, R Momenan, E Jerlhag, J A Engel, C A Hodgkinson, M Egli, M F Lopez, H C Becker, D Goldman, M Heilig, V A Ramchandani, L Leggio, P Suchankova, J Yan, M L Schwandt, B L Stangl, E C Caparelli, R Momenan, E Jerlhag, J A Engel, C A Hodgkinson, M Egli, M F Lopez, H C Becker, D Goldman, M Heilig, V A Ramchandani, L Leggio

Abstract

The hormone glucagon-like peptide-1 (GLP-1) regulates appetite and food intake. GLP-1 receptor (GLP-1R) activation also attenuates the reinforcing properties of alcohol in rodents. The present translational study is based on four human genetic association studies and one preclinical study providing data that support the hypothesis that GLP-1R may have a role in the pathophysiology of alcohol use disorder (AUD). Case-control analysis (N = 908) was performed on a sample of individuals enrolled in the National Institute on Alcohol Abuse and Alcoholism (NIAAA) intramural research program. The Study of Addiction: Genetics and Environment (SAGE) sample (N = 3803) was used for confirmation purposes. Post hoc analyses were carried out on data from a human laboratory study of intravenous alcohol self-administration (IV-ASA; N = 81) in social drinkers and from a functional magnetic resonance imaging study in alcohol-dependent individuals (N = 22) subjected to a Monetary Incentive Delay task. In the preclinical study, a GLP-1R agonist was evaluated in a mouse model of alcohol dependence to demonstrate the role of GLP-1R for alcohol consumption. The previously reported functional allele 168Ser (rs6923761) was nominally associated with AUD (P = 0.004) in the NIAAA sample, which was partially replicated in males of the SAGE sample (P = 0.033). The 168 Ser/Ser genotype was further associated with increased alcohol administration and breath alcohol measures in the IV-ASA experiment and with higher BOLD response in the right globus pallidus when receiving notification of outcome for high monetary reward. Finally, GLP-1R agonism significantly reduced alcohol consumption in a mouse model of alcohol dependence. These convergent findings suggest that the GLP-1R may be an attractive target for personalized pharmacotherapy treatment of AUD.

Figures

Figure 1
Figure 1
Comparison of risk allele frequencies between non-smoking controls, non-smoking subjects with alcohol use disorder (AUD) and smoking subjects with AUD in the LCTS study using Pearson χ2 test. Total n for the three groups were 125–130, 121–128, 202–215 for the Caucasian sample and 26–29, 47–50, 137–147 for the African American sample, respectively. *P<0.05, **P<0.01, ***P<0.005. LCTS, Laboratory of Clinical and Translational Studies.
Figure 2
Figure 2
Statistical maps for fMRI analysis contrasting the genotype groups 168Gly/Gly × 168Ser/Gly+168Ser/Ser in the rs6923761 SNP. Statistical maps (top) and ROI results (bottom) for notification of high reward (left) and its net difference with low reward (right) contrasting (168Gly/Gly)−(168Ser/Gly+168Ser/Ser) are shown; ROI localization is displayed in yellow at the axial view in the bottom. fMRI, functional magnetic resonance imaging; SNP, single-nucleotide polymorphism.
Figure 3
Figure 3
GLP-1R agonism in a mouse model of alcohol dependence. (a) AC3174 treatment #1 (alcohol intake during test cycle 5): EtOH mice consumed significantly more alcohol than CTL mice (P< 0.001). Analysis of variance (ANOVA) failed to indicate a main effect of treatment or group × treatment interaction. (b) AC3174 treatment #2 (alcohol intake during test cycle 6): there was a significant main effect of group (P<0.001), with EtOH mice consuming a greater amount of alcohol than CTL mice. ANOVA failed to indicate a main effect of treatment or group × treatment interaction. (c) AC3174 treatment #3 (alcohol intake during test cycle 7): ANOVA indicated a significant main effect of group (P<0.00001) and a significant group × treatment interaction (P<0.025). Post hoc comparisons indicated that, as expected, EtOH mice injected with vehicle consumed more alcohol than CTL mice. In addition, all doses of AC3174 significantly reduced drinking compared with the vehicle condition in EtOH mice, while AC3174 treatment did not significantly alter alcohol intake in nondependent CTL mice. Further, AC3174 treatment abolished the difference in alcohol intake between EtOH and CTL conditions. (d) Placebo (washout) test #1 (alcohol intake during test cycle 8): all mice were treated with saline (drug-washout test) to substantiate the apparent efficacy of AC3174 to reduce escalated alcohol drinking in dependent mice. ANOVA revealed a significant main effect of group (P<0.00001) and a significant group × treatment interaction (P<0.01). Post hoc comparisons supported the expected greater alcohol intake in EtOH compared with CTL mice that continued to receive vehicle. A similar profile of results was obtained in mice that received the lowest AC3174 dose (0.03 μg kg−1) in the previous test cycle. However, EtOH mice that received 0.10 or 0.30 μg kg−1 AC3174 doses in the previous test period continued to consume significantly less alcohol compared with mice that previously received vehicle, and their lower level of intake was similar to that exhibited by the corresponding CTL groups. (e) Placebo (washout) test #2 (alcohol intake during test cycle 9): ANOVA indicated a significant main effect of group (P<0.001), but no effect of treatment or an interaction between group × treatment during this second washout test period. These results indicate that after a second week of placebo (saline) treatment, elevated drinking in EtOH compared with CTL mice was restored in all the test groups. *P<0.05, significantly different from corresponding CTL group; ^P<0.05, significantly different from corresponding vehicle group.

References

    1. Larsen PJ, Holst JJ. Glucagon-related peptide 1 (GLP-1): hormone and neurotransmitter. Regul Pept. 2005;128:97–107.
    1. Pannacciulli N, Le DSNT, Salbe AD, Chen K, Reiman EM, Tataranni PA, et al. Postprandial glucagon-like peptide-1 (GLP-1) response is positively associated with changes in neuronal activity of brain areas implicated in satiety and food intake regulation in humans. Neuroimage. 2007;35:511–517.
    1. Alvarez E, Martínez MD, Roncero I, Chowen JA, García-Cuartero B, Gispert JD, et al. The expression of GLP-1 receptor mRNA and protein allows the effect of GLP-1 on glucose metabolism in the human hypothalamus and brainstem. J Neurochem. 2005;92:798–806.
    1. Merchenthaler I, Lane M, Shughrue P. Distribution of pre-pro-glucagon and glucagon-like peptide-1 receptor messenger RNAs in the rat central nervous system. J Comp Neurol. 1999;403:261–280.
    1. Alhadeff AL, Rupprecht LE, Hayes MR. 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;153:647–658.
    1. Egecioglu E, Steensland P, Fredriksson I, Feltmann K, Engel JA, Jerlhag E. The glucagon-like peptide 1 analogue Exendin-4 attenuates alcohol mediated behaviors in rodents. Psychoneuroendocrinology. 2013;38:1259–1270.
    1. Shirazi RH, Dickson SL, Skibicka KP. Gut peptide GLP-1 and its analogue, Exendin-4, decrease alcohol intake and reward. PLoS One. 2013;8:e61965.
    1. Kendler KS, Myers J. Clinical indices of familial alcohol use disorder. Alcohol Clin Exp Res. 2012;36:2126–2131.
    1. Kareken DA, Liang T, Wetherill L, Dzemidzic M, Bragulat V, Cox C, et al. A polymorphism in GABRA2 is associated with the medial frontal response to alcohol cues in an fMRI study. Alcohol Clin Exp Res. 2010;34:2169–2178.
    1. First MB, Spitzer RL, Gibbon M, Williams JBW. Structured Clinical Interview for DSM-IV-TR Axis I Disorders, Research Version, Patient Edition (SCID-I/P), vol. 1/2007 revision. Biometrics Research, New York State Psychiatric Institute: New York, NY, USA; 2002.
    1. Bierut LJ, Agrawal A, Bucholz KK, Doheny KF, Laurie C, Pugh E, et al. A genome-wide association study of alcohol dependence. Proc Natl Acad Sci USA. 2010;107:5082–5087.
    1. Yan J, Aliev F, Webb BT, Kendler KS, Williamson VS, Edenberg HJ, et al. Using genetic information from candidate gene and genome-wide association studies in risk prediction for alcohol dependence. Addict Biol. 2014;19:708–721.
    1. Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MAR, Bender D, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81:559–575.
    1. Zimmermann US, O'Connor S, Ramchandani VA. Modeling alcohol self-administration in the human laboratory. Curr Top Behav Neurosci. 2013;13:315–353.
    1. Stangl BL, Zametkin M, Hommer DW, Ramchandani VA. Alcoholism: Clinical and Experimental Research, 35th Annual Scientific Meeting of the Research Society on Alcoholism. Research Society on Alcoholism: San Francisco, CA, USA; 2012. The motivation for alcohol reward: predictors of progressive-ration intravenous (IV) alcohol self-administration in social drinkers.
    1. Ramchandani VA, Bolane J, Li TK, O'Connor S. A physiologically-based pharmacokinetic (PBPK) model for alcohol facilitates rapid BrAC clamping. Alcohol Clin Exp Res. 1999;23:617–623.
    1. Zimmermann US, O'Connor S, Ramchandani VA.Modeling alcohol self-administration in the human laboratory Sommer WH, Spanagel R.(eds). Behavioral Neurobiology of Alcohol Addiction Springer: Berlin-Heidelberg, Germany; 2013315–353.
    1. Bjork JM, Smith AR, Chen G, Hommer DW. Mesolimbic recruitment by nondrug rewards in detoxified alcoholics: effort anticipation, reward anticipation, and reward delivery. Hum Brain Mapp. 2012;33:2174–2188.
    1. Hargrove DM, Kendall ES, Reynolds JM, Lwin AN, Herich JP, Smith PA, et al. Biological activity of AC3174, a peptide analog of exendin-4. Regul Pept. 2007;141:113–119.
    1. Becker HC, Lopez MF. Increased ethanol drinking after repeated chronic ethanol exposure and withdrawal experience in C57BL/6 mice. Alcohol Clin Exp Res. 2004;28:1829–1838.
    1. Lopez MF, Becker HC. Effect of pattern and number of chronic ethanol exposures on subsequent voluntary ethanol intake in C57BL/6J mice. Psychopharmacology (Berl) 2005;181:688–696.
    1. Li J, Ji L. Adjusting multiple testing in multilocus analyses using the eigenvalues of a correlation matrix. Heredity (Edinb) 2005;95:221–227.
    1. Nyholt DR. A simple correction for multiple testing for single-nucleotide polymorphisms in linkage disequilibrium with each other. Am J Hum Genet. 2004;74:765–769.
    1. Grucza RA, Bierut LJ. Co-occurring risk factors for alcohol dependence and habitual smoking: Update on findings from the Collaborative Study on the Genetics of Alcoholism. Alcohol Res Health. 2006;29:172–178.
    1. John U, Meyer C, Rumpf H-J, Schumann A, Thyrian JR, Hapke U. Strength of the Relationship Between Tobacco Smoking, Nicotine Dependence and the Severity of Alcohol Dependence Syndrome Criteria in a Population-Based Sample. Alcohol Alcohol. 2003;38:606–612.
    1. Cox RW. AFNI: software for analysis and visualization of functional magnetic resonance neuroimages. Comput Biomed Res. 1996;29:162–173.
    1. Chen G, Adleman NE, Saad ZS, Leibenluft E, Cox RW. Applications of multivariate modeling to neuroimaging group analysis: A comprehensive alternative to univariate general linear model. Neuroimage. 2014;99:571–588.
    1. Sobell LC, Sobell MB.Timeline follow-back Litten RZ, Allen JP.(eds). Measuring Alcohol Consumption Humana Press: Totowa, NJ, USA; 199241–72.
    1. Baggio LL, Drucker DJ. Biology of Incretins: GLP-1 and GIP. Gastroenterology. 2007;132:2131–2157.
    1. Dailey MJ, Moran TH. Glucagon-like peptide 1 and appetite. Trends Endocrinol Metab. 2013;24:85–91.
    1. Baumgartner I, Pacheco-López G, Rüttimann EB, Arnold M, Asarian L, Langhans W, et al. Hepatic-portal vein infusions of glucagon-like peptide-1 reduce meal size and increase c-Fos expression in the nucleus tractus solitarii, area postrema and central nucleus of the amygdala in rats. J Neuroendocrinol. 2010;22:557–563.
    1. Parker JA, McCullough KA, Field BC, Minnion JS, Martin NM, Ghatei MA, et al. Glucagon and GLP-1 inhibit food intake and increase c-fos expression in similar appetite regulating centres in the brainstem and amygdala. Int J Obes (Lond) 2013;37:1391–1398.
    1. Koole C, Wootten D, Simms J, Valant C, Miller LJ, Christopoulos A, et al. Polymorphism and ligand dependent changes in human glucagon-like peptide-1 receptor (GLP-1R) function: allosteric rescue of loss of function mutation. Mol Pharmacol. 2011;80:486–497.
    1. Sathananthan A, Man CD, Micheletto F, Zinsmeister AR, Camilleri M, Giesler PD, et al. Common genetic variation in GLP1R and insulin secretion in response to exogenous GLP-1 in nondiabetic subjects: a pilot study. Diabetes Care. 2010;33:2074–2076.
    1. de Luis DA, Aller R, de la Fuente B, Primo D, Conde R, Izaola O, et al. Relation of the rs6923761 gene variant in glucagon-like peptide 1 receptor with weight, cardiovascular risk factor, and serum adipokine levels in obese female subjects. J Clin Lab Anal. 2014;29:100–105.
    1. Ramsey TL, Brennan MD. Glucagon-like peptide 1 receptor (GLP1R) haplotypes correlate with altered response to multiple antipsychotics in the CATIE trial. Schizophr Res. 2014;160:73–79.
    1. Sheikh HI, Dougherty LR, Hayden EP, Klein DN, Singh SM. Glucagon-like peptide-1 receptor gene polymorphism (Leu260Phe) is associated with morning cortisol in preschoolers. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:980–983.
    1. Heilig M, Goldman D, Berrettini W, O'Brien CP. Pharmacogenetic approaches to the treatment of alcohol addiction. Nat Rev Neurosci. 2011;12:670–684.
    1. Prescott CA, Aggen SH, Kendler KS. Sex differences in the sources of genetic liability to alcohol abuse and dependence in a population-based sample of U.S. twins. Alcohol Clin Exp Res. 1999;23:1136–1144.
    1. Prescott CA, Kendler KS. Genetic and environmental contributions to alcohol abuse and dependence in a population-based sample of male twins. Am J Psychiatry. 1999;156:34–40.
    1. Schlaepfer IR, Hoft NR, Ehringer MA. The genetic components of alcohol and nicotine co-addiction: from genes to behavior. Curr Drug Abuse Rev. 2008;1:124–134.
    1. Egecioglu E, Engel JA, Jerlhag E. The glucagon-like peptide 1 analogue exendin-4 attenuates the nicotine-induced locomotor stimulation, accumbal dopamine release, conditioned place preference as well as the expression of locomotor sensitization in mice. PLoS One. 2013;8:e77284.
    1. Hurt RD, Offord KP, Croghan IT, Gomez-Dahl L, Kottke TE, Morse RM, et al. Mortality following inpatient addictions treatment. Role of tobacco use in a community-based cohort. JAMA. 1996;275:1097–1103.
    1. Hong S, Hikosaka O. The globus pallidus sends reward-related signals to the lateral habenula. Neuron. 2008;60:720–729.
    1. Calder AJ, Beaver JD, Davis MH, van Ditzhuijzen J, Keane J, Lawrence AD. Disgust sensitivity predicts the insula and pallidal response to pictures of disgusting foods. Eur J Neurosci. 2007;25:3422–3428.
    1. Haber SN, Wolfe DP, Groenewegen HJ. The relationship between ventral striatal efferent fibers and the distribution of peptide-positive woolly fibers in the forebrain of the rhesus monkey. Neuroscience. 1990;39:323–338.
    1. Schott BH, Minuzzi L, Krebs RM, Elmenhorst D, Lang M, Winz OH, et al. Mesolimbic functional magnetic resonance imaging activations during reward anticipation correlate with reward-related ventral striatal dopamine release. J Neurosci. 2008;28:14311–14319.
    1. Erreger K, Davis AR, Poe AM, Greig NH, Stanwood GD, Galli A. Exendin-4 decreases amphetamine-induced locomotor activity. Physiol Behav. 2012;106:574–578.
    1. Mietlicki-Baase EG, Ortinski PI, Rupprecht LE, Olivos DR, Alhadeff AL, Pierce RC, et al. The food intake-suppressive effects of glucagon-like peptide-1 receptor signaling in the ventral tegmental area are mediated by AMPA/kainate receptors. Am J Physiol Endocrinol Metab. 2013;305:E1367–E1374.
    1. Volkow ND, Wang GJ, Maynard L, Fowler JS, Jayne B, Telang F, et al. Effects of alcohol detoxification on dopamine D2 receptors in alcoholics: a preliminary study. Psychiatry Res. 2002;116:163–172.
    1. Sen N. Epigenetic regulation of memory by acetylation and methylation of chromatin: implications in neurological disorders, aging, and addiction. Neuromolecular Med. 2014;17:97–110.

Source: PubMed

3
Abonnieren