Differential association between the GLP1R gene variants and brain functional connectivity according to the severity of alcohol use

Mehdi Farokhnia, Samantha J Fede, Erica N Grodin, Brittney D Browning, Madeline E Crozier, Melanie L Schwandt, Colin A Hodgkinson, Reza Momenan, Lorenzo Leggio, Mehdi Farokhnia, Samantha J Fede, Erica N Grodin, Brittney D Browning, Madeline E Crozier, Melanie L Schwandt, Colin A Hodgkinson, Reza Momenan, Lorenzo Leggio

Abstract

Growing evidence suggests that the glucagon-like peptide-1 (GLP-1) system is involved in mechanisms underlying alcohol seeking and consumption. Accordingly, the GLP-1 receptor (GLP-1R) has begun to be studied as a potential pharmacotherapeutic target for alcohol use disorder (AUD). The aim of this study was to investigate the association between genetic variation at the GLP-1R and brain functional connectivity, according to the severity of alcohol use. Participants were 181 individuals categorized as high-risk (n = 96) and low-risk (n = 85) alcohol use, according to their AUD identification test (AUDIT) score. Two uncommon single nucleotide polymorphisms (SNPs), rs6923761 and rs1042044, were selected a priori for this study because they encode amino-acid substitutions with putative functional consequences on GLP-1R activity. Genotype groups were based on the presence of the variant allele for each of the two GLP-1R SNPs of interest [rs6923761: AA + AG (n = 65), GG (n = 116); rs1042044: AA + AC (n = 114), CC (n = 67)]. Resting-state functional MRI data were acquired for 10 min and independent component (IC) analysis was conducted. Multivariate analyses of covariance (MANCOVA) examined the interaction between GLP-1R genotype group and AUDIT group on within- and between-network connectivity. For rs6923761, three ICs showed significant genotype × AUDIT interaction effects on within-network connectivity: two were mapped onto the anterior salience network and one was mapped onto the visuospatial network. For rs1042044, four ICs showed significant interaction effects on within-network connectivity: three were mapped onto the dorsal default mode network and one was mapped onto the basal ganglia network. For both SNPs, post-hoc analyses showed that in the group carrying the variant allele, high versus low AUDIT was associated with stronger within-network connectivity. No significant effects on between-network connectivity were found. In conclusion, genetic variation at the GLP-1R was differentially associated with brain functional connectivity in individuals with low versus high severity of alcohol use. Significant findings in the salience and default mode networks are particularly relevant, given their role in the neurobiology of AUD and addictive behaviors.

Conflict of interest statement

The authors declare no competing interests.

© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.

Figures

Figure 1
Figure 1
Normalized within-network connectivity estimates for significant interactions between AUDIT group and rs6923761 genotype. Top panels (brain images) include the component T-map extracted during the ICA processing step; colors represent T-max values. High-AUDIT: AUDIT total score ≥ 8 (n = 96); Low-AUDIT: AUDIT total score n = 85). A is the risk allele; A-allele carriers: AA + AG (n = 65); Non-A-allele carriers: GG (n = 116). IC35 (insula; A) and IC47 (frontal/acc/putamen; B) were mapped onto the anterior salience network; IC21 (insula/occipital/mpfc; C) was mapped onto the visuospatial network.
Figure 2
Figure 2
Normalized within-network connectivity estimates for significant interactions between AUDIT group and rs1042044 genotype. Top panels (brain images) include the component T-map extracted during the ICA processing step; colors represent T-max values. High-AUDIT: AUDIT total score ≥ 8 (n = 96); Low-AUDIT: AUDIT total score < 8 (n = 85). A is the risk allele; A-allele carriers: AA + AC (n = 114); Non-A-allele carriers: CC (n = 67). IC8 (hypothalamus/brainstem; A), IC45 (thalamus/3rd ventricle; B), and IC54 (pcc/tpj/occipital/mpfc; C) were mapped onto the dorsal default mode network; IC46 (thalamus/brainstem; D) was mapped onto the basal ganglia network.

References

    1. GBD, The global burden of disease attributable to alcohol and drug use in 195 countries and territories, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Psychiatry, 2018. 5(12): pp. 987–1012.
    1. Heilig M, et al. Addiction as a brain disease revised: Why it still matters, and the need for consilience. Neuropsychopharmacology. 2021;46:1715. doi: 10.1038/s41386-020-00950-y.
    1. Ray LA, et al. The future of translational research on alcohol use disorder. Addict. Biol. 2021;26(2):e12903. doi: 10.1111/adb.12903.
    1. Volkow ND, et al. Food and drug reward: Overlapping circuits in human obesity and addiction. Curr. Top Behav. Neurosci. 2012;11:1–24.
    1. Faulkner ML, Momenan R, Leggio L. A neuroimaging investigation into the role of peripheral metabolic biomarkers in the anticipation of reward in alcohol use. Drug Alcohol Depend. 2021;221:108638. doi: 10.1016/j.drugalcdep.2021.108638.
    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. 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(2):647–658. doi: 10.1210/en.2011-1443.
    1. Kanoski SE, et al. Peripheral and central GLP-1 receptor populations mediate the anorectic effects of peripherally administered GLP-1 receptor agonists, liraglutide and exendin-4. Endocrinology. 2011;152(8):3103–3112. doi: 10.1210/en.2011-0174.
    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, 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(2):261–280. doi: 10.1002/(SICI)1096-9861(19990111)403:2<261::AID-CNE8>;2-5.
    1. Alvarez E, 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(4):798–806. doi: 10.1111/j.1471-4159.2004.02914.x.
    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. Alhadeff AL, et al. Glucagon-like Peptide-1 receptor signaling in the lateral parabrachial nucleus contributes to the control of food intake and motivation to feed. Neuropsychopharmacology. 2014;39(9):2233–2243. doi: 10.1038/npp.2014.74.
    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. Neurosc. 2012;32(14):4812–4820. doi: 10.1523/JNEUROSCI.6326-11.2012.
    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 doi: 10.3389/fnbeh.2020.614884.
    1. Ghosal S, Myers B, Herman JP. Role of central glucagon-like peptide-1 in stress regulation. Physiol. Behav. 2013;122:201–207. doi: 10.1016/j.physbeh.2013.04.003.
    1. Holt MK, Trapp S. The physiological role of the brain GLP-1 system in stress. Cogent Biol. 2016;2(1):1229086–1229086. doi: 10.1080/23312025.2016.1229086.
    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. 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. Vallöf D, Vestlund J, Jerlhag E. Glucagon-like peptide-1 receptors within the nucleus of the solitary tract regulate alcohol-mediated behaviors in rodents. Neuropharmacology. 2019;149:124–132. doi: 10.1016/j.neuropharm.2019.02.020.
    1. Vallöf D, et al. The glucagon-like peptide 1 receptor agonist liraglutide attenuates the reinforcing properties of alcohol in rodents. Addict. Biol. 2016;21(2):422–437. doi: 10.1111/adb.12295.
    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(4):e61965. doi: 10.1371/journal.pone.0061965.
    1. Sirohi S, et al. Central & peripheral glucagon-like peptide-1 receptor signaling differentially regulate addictive behaviors. Physiol. Behav. 2016;161:140–144. doi: 10.1016/j.physbeh.2016.04.013.
    1. Sørensen G, Caine SB, Thomsen M. Effects of the GLP-1 agonist exendin-4 on intravenous ethanol self-administration in mice. Alcohol. Clin. Exp. Res. 2016;40(10):2247–2252. doi: 10.1111/acer.13199.
    1. Thomsen M, et al. The glucagon-like peptide 1 receptor agonist Exendin-4 decreases relapse-like drinking in socially housed mice. Pharmacol. Biochem. Behav. 2017;160:14–20. doi: 10.1016/j.pbb.2017.07.014.
    1. Vallöf D, Kalafateli AL, Jerlhag E. Long-term treatment with a glucagon-like peptide-1 receptor agonist reduces ethanol intake in male and female rats. Transl. Psychiatry. 2020;10(1):238. doi: 10.1038/s41398-020-00923-1.
    1. Dixon TN, McNally GP, Ong ZY. 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. Marty VN, et al. Long-acting glucagon-like Peptide-1 receptor agonists suppress voluntary alcohol intake in male wistar rats. Front. Neurosci. 2020;14:599646. doi: 10.3389/fnins.2020.599646.
    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:e583. doi: 10.1038/tp.2015.68.
    1. Jerlhag E. GLP-1 signaling and alcohol-mediated behaviors; Preclinical and clinical evidence. Neuropharmacology. 2018;136(Pt B):343–349. doi: 10.1016/j.neuropharm.2018.01.013.
    1. De Silva A, 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(5):700–706. doi: 10.1016/j.cmet.2011.09.010.
    1. Pannacciulli N, 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(2):511–517. doi: 10.1016/j.neuroimage.2006.12.035.
    1. Heni M, et al. Differential effect of glucose ingestion on the neural processing of food stimuli in lean and overweight adults. Hum. Brain Mapp. 2014;35(3):918–928. doi: 10.1002/hbm.22223.
    1. Schloegl H, et al. Peptide hormones regulating appetite–focus on neuroimaging studies in humans. Diabetes Metab. Res. Rev. 2011;27(2):104–112. doi: 10.1002/dmrr.1154.
    1. Sathananthan A, 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(9):2074–2076. doi: 10.2337/dc10-0200.
    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(1–3):73–79. doi: 10.1016/j.schres.2014.09.038.
    1. Koole C, 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(3):486–497. doi: 10.1124/mol.111.072884.
    1. Javorský M, et al. A missense variant in GLP1R gene is associated with the glycaemic response to treatment with gliptins. Diabetes Obes. Metab. 2016;18(9):941–944. doi: 10.1111/dom.12682.
    1. Sheikh HI, et al. Glucagon-like peptide-1 receptor gene polymorphism (Leu260Phe) is associated with morning cortisol in preschoolers. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2010;34(6):980–983. doi: 10.1016/j.pnpbp.2010.05.007.
    1. de Luis DA, et al. Role of the rs6923761 gene variant in glucagon-like peptide 1 receptor gene on cardiovascular risk factors and weight loss after biliopancreatic diversion surgery. Ann. Nutr. Metab. 2014;65(4):259–263. doi: 10.1159/000365975.
    1. de Luis DA, et al. Role of rs6923761 gene variant in glucagon-like peptide 1 receptor in basal GLP-1 levels, cardiovascular risk factor and serum adipokine levels in naïve type 2 diabetic patients. J. Endocrinol. Invest. 2015;38(2):143–147. doi: 10.1007/s40618-014-0161-y.
    1. de Luis DA, 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. 2015;29(2):100–105. doi: 10.1002/jcla.21735.
    1. Fede SJ, et al. Resting state connectivity best predicts alcohol use severity in moderate to heavy alcohol users. Neuroimage Clin. 2019;22:101782. doi: 10.1016/j.nicl.2019.101782.
    1. Saunders JB, et al. Development of the alcohol use disorders identification test (AUDIT): WHO collaborative project on early detection of persons with harmful alcohol consumption-II. Addiction. 1993;88(6):791–804. doi: 10.1111/j.1360-0443.1993.tb02093.x.
    1. Allen JP, et al. A review of research on the alcohol use disorders identification test (AUDIT) Alcohol. Clin. Exp. Res. 1997;21(4):613–619. doi: 10.1111/j.1530-0277.1997.tb03811.x.
    1. Conigrave KM, Hall WD, Saunders JB. The AUDIT questionnaire: Choosing a cut-off score. Addiction. 1995;90(10):1349–1356. doi: 10.1111/j.1360-0443.1995.tb03552.x.
    1. Hodgkinson CA, et al. Addictions biology: Haplotype-based analysis for 130 candidate genes on a single array. Alcohol. Alcohol. 2008;43(5):505–515. doi: 10.1093/alcalc/agn032.
    1. Cox RW. AFNI: Software for analysis and visualization of functional magnetic resonance neuroimages. Comput. Biomed. Res. 1996;29(3):162–173. doi: 10.1006/cbmr.1996.0014.
    1. Allen EA, et al. A baseline for the multivariate comparison of resting-state networks. Front. Syst. Neurosci. 2011;5:2.
    1. Himberg, J. Hyvarinen, A. Icasso: software for investigating the reliability of ICA estimates by clustering and visualization. In 2003 IEEE XIII Workshop on Neural Networks for Signal Processing (IEEE Cat. No. 03TH8718). 2003. IEEE.
    1. Antonsen KK, et al. Does glucagon-like peptide-1 (GLP-1) receptor agonist stimulation reduce alcohol intake in patients with alcohol dependence: Study protocol of a randomised, double-blinded, placebo-controlled clinical trial. BMJ Open. 2018;8(7):e019562. doi: 10.1136/bmjopen-2017-019562.
    1. Hashimoto R, et al. Imaging genetics and psychiatric disorders. Curr. Mol. Med. 2015;15(2):168–175. doi: 10.2174/1566524015666150303104159.
    1. Gerber AJ, et al. Imaging genetics. J. Am. Acad. Child Adolesc. Psychiatry. 2009;48(4):356–361. doi: 10.1097/CHI.0b013e31819aad07.
    1. Mascarell Maričić L, et al. The IMAGEN study: A decade of imaging genetics in adolescents. Mol Psychiatry. 2020;25(11):2648–2671. doi: 10.1038/s41380-020-0822-5.
    1. Zhu X, et al. Resting-state functional connectivity and presynaptic monoamine signaling in Alcohol Dependence. Hum. Brain Mapp. 2015;36(12):4808–4818. doi: 10.1002/hbm.22951.
    1. Koob GF, et al. Addiction as a stress surfeit disorder. Neuropharmacology. 2014;2014(76):370–82. doi: 10.1016/j.neuropharm.2013.05.024.
    1. Menon V, Uddin LQ. Saliency, switching, attention and control: a network model of insula function. Brain Struct. Funct. 2010;214(5):655–667. doi: 10.1007/s00429-010-0262-0.
    1. Grodin EN, et al. Structural deficits in salience network regions are associated with increased impulsivity and compulsivity in alcohol dependence. Drug Alcohol. Depend. 2017;179:100–108. doi: 10.1016/j.drugalcdep.2017.06.014.
    1. Galandra C, et al. Salience network structural integrity predicts executive impairment in alcohol use disorders. Sci. Rep. 2018;8(1):14481. doi: 10.1038/s41598-018-32828-x.
    1. Muscogiuri G, et al. Glucagon-like Peptide-1 and the central/peripheral nervous system: crosstalk in diabetes. Trends Endocrinol. Metab. 2017;28(2):88–103. doi: 10.1016/j.tem.2016.10.001.
    1. Yaribeygi H, et al. GLP-1 mimetics and cognition. Life Sci. 2021;264:118645. doi: 10.1016/j.lfs.2020.118645.
    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. de Graaf TA, et al. Brain network dynamics underlying visuospatial judgment: An FMRI connectivity study. J. Cogn. Neurosci. 2010;22(9):2012–2026. doi: 10.1162/jocn.2009.21345.
    1. Monnig MA, et al. Diffusion tensor imaging of white matter networks in individuals with current and remitted alcohol use disorders and comorbid conditions. Psychol. Addict Behav. 2013;27(2):455–465. doi: 10.1037/a0027168.
    1. Fukushima S, et al. Behavioural cue reactivity to alcohol-related and non-alcohol-related stimuli among individuals with alcohol use disorder: An fMRI study with a visual task. PLoS ONE. 2020;15(7):e0229187. doi: 10.1371/journal.pone.0229187.
    1. Khan DM, et al. Effective connectivity for default mode network analysis of alcoholism. Brain Connect. 2021;11(1):12–29. doi: 10.1089/brain.2019.0721.
    1. Suarez AN, Noble EE, Kanoski SE. Regulation of memory function by feeding-relevant biological systems: Following the breadcrumbs to the hippocampus. Front. Mol. Neurosci. 2019;12:101. doi: 10.3389/fnmol.2019.00101.
    1. Spreng RN, et al. Default network activity, coupled with the frontoparietal control network, supports goal-directed cognition. Neuroimage. 2010;53(1):303–317. doi: 10.1016/j.neuroimage.2010.06.016.
    1. Kamarajan C, et al. Random forest classification of alcohol use disorder using EEG source functional connectivity, neuropsychological functioning, and impulsivity measures. Behav. Sci. (Basel) 2020;10(3):62. doi: 10.3390/bs10030062.
    1. Fang X, et al. Effects of moderate alcohol levels on default mode network connectivity in heavy drinkers. Alcohol. Clin. Exp. Res. 2021;45(5):1039–1050. doi: 10.1111/acer.14602.
    1. Song Z, et al. Abnormal functional connectivity and effective connectivity between the default mode network and attention networks in patients with alcohol-use disorder. Acta Radiol. 2021;62(2):251–259. doi: 10.1177/0284185120923270.
    1. van Duinkerken E, et al. Cerebral effects of glucagon-like peptide-1 receptor blockade before and after Roux-en-Y gastric bypass surgery in obese women: A proof-of-concept resting-state functional MRI study. Diabetes Obes. Metab. 2021;23(2):415–424. doi: 10.1111/dom.14233.
    1. Lanciego JL, Luquin N, Obeso JA. Functional neuroanatomy of the basal ganglia. Cold Spring Harb. Perspect. Med. 2012;2(12):a009621. doi: 10.1101/cshperspect.a009621.
    1. Lovinger DM, Alvarez VA. Alcohol and basal ganglia circuitry: Animal models. Neuropharmacology. 2017;122:46–55. doi: 10.1016/j.neuropharm.2017.03.023.
    1. Modell JG, Mountz JM, Beresford TP. Basal ganglia/limbic striatal and thalamocortical involvement in craving and loss of control in alcoholism. J. Neuropsychiatry Clin. Neurosci. 1990;2(2):123–144. doi: 10.1176/jnp.2.2.123.
    1. Mora F, et al. Selective release of glutamine and glutamic acid produced by perfusion of GLP-1 (7–36) amide in the basal ganglia of the conscious rat. Brain Res. Bull. 1992;29(3–4):359–361. doi: 10.1016/0361-9230(92)90068-9.
    1. Rothman KJ. No adjustments are needed for multiple comparisons. Epidemiology. 1990;1(1):43–46. doi: 10.1097/00001648-199001000-00010.

Source: PubMed

3
Abonner