Skip to main content

Neural Circuitries and Alcohol Use Disorder: Cutting Corners in the Cycle

  • Chapter
  • First Online:
Current Topics in Behavioral Neurosciences

Abstract

An implicit tenet of the alcohol use disorder (AUD) research field is that knowledge of how alcohol interacts with the brain is critical to the development of an understanding of vulnerability to AUD and treatment approaches. Gaining this understanding requires the mapping of brain function critical to specific components of this heterogeneous disorder. Early approaches in humans and animal models focused on the determination of specific brain regions sensitive to alcohol action and their participation in AUD-relevant behaviors. Broadly speaking, this research has focused on three domains, Binge/Intoxication, Negative Affect/Withdrawal, and Preoccupation/Anticipation, with a number of regions identified as participating in each. With the generational advances in technologies that the field of neuroscience has undergone over the last two decades, this focus has shifted to a circuit-based analysis. A wealth of new data has sharpened the field’s focus on the specific roles of the interconnectivity of multiple brain regions in AUD and AUD-relevant behaviors, as well as demonstrating that the three major domains described above have much fuzzier edges than originally thought.

In this chapter, we very briefly review brain regions previously implicated in aspects of AUD-relevant behavior from animal model research. Next, we move to a more in-depth overview of circuit-based approaches, and the utilization of these approaches in current AUD research.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Institutional subscriptions

References

  • Abrahao KP, Chancey JH, Chan CS, Lovinger DM (2017) Ethanol-sensitive pacemaker neurons in the mouse external Globus pallidus. Neuropsychopharmacology 42(5):1070–1081

    Google Scholar 

  • Arinze I, Moorman DE (2020) Selective impact of lateral orbitofrontal cortex inactivation on reinstatement of alcohol seeking in male Long-Evans rats. Neuropharmacology 168:108007

    Google Scholar 

  • Arizzi-LaFrance MN, Correa M, Aragon CM, Salamone JD (2006) Motor stimulant effects of ethanol injected into the substantia nigra pars reticulata: importance of catalase-mediated metabolism and the role of acetaldehyde. Neuropsychopharmacology 31(5):997–1008

    Google Scholar 

  • Armand EJ, Li J, Xie F, Luo C, Mukamel EA (2021) Single-cell sequencing of brain cell transcriptomes and epigenomes. Neuron 109(1):11–26

    Google Scholar 

  • Avegno EM, Gilpin NW (2022) Reciprocal midbrain-extended amygdala circuit activity in preclinical models of alcohol use and misuse. Neuropharmacology 202:108856

    Google Scholar 

  • Avegno EM, Kasten CR, Snyder WB 3rd, Kelley LK, Lobell TD, Templeton TJ, Constans M, Wills TA, Middleton JW, Gilpin NW (2021) Alcohol dependence activates ventral tegmental area projections to central amygdala in male mice and rats. Addict Biol 26(4):e12990

    Google Scholar 

  • Bianchi PC, Carneiro de Oliveira PE, Palombo P, Leao RM, Cogo-Moreira H, Planeta CDS, Cruz FC (2018) Functional inactivation of the orbitofrontal cortex disrupts context-induced reinstatement of alcohol seeking in rats. Drug Alcohol Depend 186:102–112

    Google Scholar 

  • Brodie MS, Shefner SA, Dunwiddie TV (1990) Ethanol increases the firing rate of dopamine neurons of the rat ventral tegmental area in vitro. Brain Res 508(1):65–69

    Google Scholar 

  • Burnham NW, Chaimowitz CN, Vis CC, Segantine Dornellas AP, Navarro M, Thiele TE (2021) Lateral hypothalamus-projecting noradrenergic locus coeruleus pathway modulates binge-like ethanol drinking in male and female TH-ires-cre mice. Neuropharmacology 196:108702

    Google Scholar 

  • Cassataro D, Bergfeldt D, Malekian C, Van Snellenberg JX, Thanos PK, Fishell G, Sjulson L (2014) Reverse pharmacogenetic modulation of the nucleus accumbens reduces ethanol consumption in a limited access paradigm. Neuropsychopharmacology 39(2):283–290

    Google Scholar 

  • Centanni SW, Bedse G, Patel S, Winder DG (2019a) Driving the downward spiral: alcohol-induced dysregulation of extended Amygdala circuits and negative affect. Alcohol Clin Exp Res 43(10):2000–2013

    Google Scholar 

  • Centanni SW, Morris BD, Luchsinger JR, Bedse G, Fetterly TL, Patel S, Winder DG (2019b) Endocannabinoid control of the insular-bed nucleus of the stria terminalis circuit regulates negative affective behavior associated with alcohol abstinence. Neuropsychopharmacology 44(3):526–537

    Google Scholar 

  • Cheng Y, Huang CCY, Ma T, Wei X, Wang X, Lu J, Wang J (2017) Distinct synaptic strengthening of the striatal direct and indirect pathways drives alcohol consumption. Biol Psychiatry 81(11):918–929

    Google Scholar 

  • Corbit LH, Nie H, Janak PH (2012) Habitual alcohol seeking: time course and the contribution of subregions of the dorsal striatum. Biol Psychiatry 72(5):389–395

    Google Scholar 

  • Craig AD (2009) How do you feel – now? The anterior insula and human awareness. Nat Rev Neurosci 10(1):59–70

    Google Scholar 

  • Criswell HE, Simson PE, Knapp DJ, Devaud LL, McCown TJ, Duncan GE, Morrow AL, Breese GR (1995) Effect of zolpidem on gamma-aminobutyric acid (GABA)-induced inhibition predicts the interaction of ethanol with GABA on individual neurons in several rat brain regions. J Pharmacol Exp Ther 273(1):526–536

    Google Scholar 

  • Dayas CV, Liu X, Simms JA, Weiss F (2007) Distinct patterns of neural activation associated with ethanol seeking: effects of naltrexone. Biol Psychiatry 61(8):979–989

    Google Scholar 

  • de Guglielmo G, Crawford E, Kim S, Vendruscolo LF, Hope BT, Brennan M, Cole M, Koob GF, George O (2016) Recruitment of a neuronal ensemble in the central nucleus of the Amygdala is required for alcohol dependence. J Neurosci 36(36):9446–9453

    Google Scholar 

  • de Guglielmo G, Kallupi M, Pomrenze MB, Crawford E, Simpson S, Schweitzer P, Koob GF, Messing RO, George O (2019) Inactivation of a CRF-dependent amygdalofugal pathway reverses addiction-like behaviors in alcohol-dependent rats. Nat Commun 10(1):1238

    Google Scholar 

  • De Oliveira Sergio T, Lei K, Kwok C, Ghotra S, Wegner SA, Walsh M, Waal J, Darevsky D, Hopf FW (2021) The role of anterior insula-brainstem projections and alpha-1 noradrenergic receptors for compulsion-like and alcohol-only drinking. Neuropsychopharmacology 46(11):1918–1926

    Google Scholar 

  • Di Chiara G, Imperato A (1988) Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proc Natl Acad Sci U S A 85(14):5274–5278

    Google Scholar 

  • Domi E, Domi A, Adermark L, Heilig M, Augier E (2021) Neurobiology of alcohol seeking behavior. J Neurochem 157(5):1585–1614

    Google Scholar 

  • Dong C, Zheng Y, Long-Iyer K, Wright EC, Li Y, Tian L (2022) Fluorescence imaging of neural activity, neurochemical dynamics, and drug-specific receptor conformation with genetically encoded sensors. Annu Rev Neurosci 45:273–294

    Google Scholar 

  • Dornellas APS, Burnham NW, Luhn KL, Petruzzi MV, Thiele TE, Navarro M (2021) Activation of locus coeruleus to rostromedial tegmental nucleus (RMTg) noradrenergic pathway blunts binge-like ethanol drinking and induces aversive responses in mice. Neuropharmacology 199:108797

    Google Scholar 

  • Droutman V, Bechara A, Read SJ (2015a) Roles of the different sub-regions of the insular cortex in various phases of the decision-making process. Front Behav Neurosci 9:309

    Google Scholar 

  • Droutman V, Read SJ, Bechara A (2015b) Revisiting the role of the insula in addiction. Trends Cogn Sci 19(7):414–420

    Google Scholar 

  • Felipe JM, Palombo P, Bianchi PC, Zaniboni CR, AnĂ©sio A, Yokoyama TS, Engi SA, Carneiro-de-Oliveira PE, Planeta CDS, LeĂŁo RM, Cruz FC (2021) Dorsal hippocampus plays a causal role in context-induced reinstatement of alcohol-seeking in rats. Behav Brain Res 398:112978

    Google Scholar 

  • Feltenstein MW, See RE, Fuchs RA (2021) Neural substrates and circuits of drug addiction. Cold Spring Harb Perspect Med 11(4)

    Google Scholar 

  • Fetterly TL, Basu A, Nabit BP, Awad E, Williford KM, Centanni SW, Matthews RT, Silberman Y, Winder DG (2019) alpha2A-adrenergic receptor activation decreases parabrachial nucleus excitatory drive onto BNST CRF neurons and reduces their activity in vivo. J Neurosci 39(3):472–484

    Google Scholar 

  • Fuchs RA, Evans KA, Ledford CC, Parker MP, Case JM, Mehta RH, See RE (2005) The role of the dorsomedial prefrontal cortex, basolateral amygdala, and dorsal hippocampus in contextual reinstatement of cocaine seeking in rats. Neuropsychopharmacology 30(2):296–309

    Google Scholar 

  • Gatto GJ, McBride WJ, Murphy JM, Lumeng L, Li TK (1994) Ethanol self-infusion into the ventral tegmental area by alcohol-preferring rats. Alcohol 11(6):557–564

    Google Scholar 

  • George O, Hope BT (2017) Cortical and amygdalar neuronal ensembles in alcohol seeking, drinking and withdrawal. Neuropharmacology 122:107–114

    Google Scholar 

  • Gessa GL, Muntoni F, Collu M, Vargiu L, Mereu G (1985) Low doses of ethanol activate dopaminergic neurons in the ventral tegmental area. Brain Res 348(1):201–203

    Google Scholar 

  • Gremel CM, Cunningham CL (2008) Roles of the nucleus accumbens and amygdala in the acquisition and expression of ethanol-conditioned behavior in mice. J Neurosci 28(5):1076–1084

    Google Scholar 

  • Hansson AC, Rimondini R, Neznanova O, Sommer WH, Heilig M (2008) Neuroplasticity in brain reward circuitry following a history of ethanol dependence. Eur J Neurosci 27(8):1912–1922

    Google Scholar 

  • Heidenreich M, Zhang F (2016) Applications of CRISPR-Cas systems in neuroscience. Nat Rev Neurosci 17(1):36–44

    Google Scholar 

  • Hernandez JS, Binette AN, Rahman T, Tarantino JD, Moorman DE (2020) Chemogenetic inactivation of orbitofrontal cortex decreases Cue-induced reinstatement of ethanol and sucrose seeking in male and female Wistar rats. Alcohol Clin Exp Res 44(9):1769–1782

    Google Scholar 

  • Herring BE, Mayfield RD, Camp MC, Alcantara AA (2004) Ethanol-induced Fos immunoreactivity in the extended amygdala and hypothalamus of the rat brain: focus on cholinergic interneurons of the nucleus accumbens. Alcohol Clin Exp Res 28(4):588–597

    Google Scholar 

  • Hitzemann B, Hitzemann R (1997) Genetics ethanol and the Fos response: a comparison of the C57BL/6J and DBA/2J inbred mouse strains. Alcohol Clin Exp Res 21(8):1497–1507

    Google Scholar 

  • Hodge CW, Chappelle AM, Samson HH (1995) GABAergic transmission in the nucleus accumbens is involved in the termination of ethanol self-administration in rats. Alcohol Clin Exp Res 19(6):1486–1493

    Google Scholar 

  • Hodge CW, Haraguchi M, Erickson H, Samson HH (1993) Ventral tegmental microinjections of quinpirole decrease ethanol and sucrose-reinforced responding. Alcohol Clin Exp Res 17(2):370–375

    Google Scholar 

  • Hodge CW, Samson HH, Chappelle AM (1997) Alcohol self-administration: further examination of the role of dopamine receptors in the nucleus accumbens. Alcohol Clin Exp Res 21(6):1083–1091

    Google Scholar 

  • Hoffman PL, Rabe CS, Moses F, Tabakoff B (1989) N-methyl-D-aspartate receptors and ethanol: inhibition of calcium flux and cyclic GMP production. J Neurochem 52(6):1937–1940

    Google Scholar 

  • Holleran KM, Wilson HH, Fetterly TL, Bluett RJ, Centanni SW, Gilfarb RA, Rocco LE, Patel S, Winder DG (2016) Ketamine and MAG lipase inhibitor-dependent reversal of evolving depressive-like behavior during forced abstinence from alcohol drinking. Neuropsychopharmacology 41(8):2062–2071

    Google Scholar 

  • Hong SI, Kang S, Chen JF, Choi DS (2019) Indirect medium spiny neurons in the dorsomedial striatum regulate ethanol-containing conditioned reward seeking. J Neurosci 39(36):7206–7217

    Google Scholar 

  • Hosp F, Mann M (2017) A primer on concepts and applications of proteomics in neuroscience. Neuron 96(3):558–571

    Google Scholar 

  • Janak PH, Chaudhri N (2010) The potent effect of environmental context on relapse to alcohol-seeking after extinction. Open Addict J 3:76–87

    Google Scholar 

  • Jaramillo AA, Agan VE, Makhijani VH, Pedroza S, McElligott ZA, Besheer J (2018a) Functional role for suppression of the insular-striatal circuit in modulating interoceptive effects of alcohol. Addict Biol 23(5):1020–1031

    Google Scholar 

  • Jaramillo AA, Randall PA, Frisbee S, Fisher KR, Besheer J (2015) Activation of mGluR2/3 following stress hormone exposure restores sensitivity to alcohol in rats. Alcohol 49(6):525–532

    Google Scholar 

  • Jaramillo AA, Van Voorhies K, Randall PA, Besheer J (2018b) Silencing the insular-striatal circuit decreases alcohol self-administration and increases sensitivity to alcohol. Behav Brain Res 348:74–81

    Google Scholar 

  • Jasinska AJ, Stein EA, Kaiser J, Naumer MJ, Yalachkov Y (2014) Factors modulating neural reactivity to drug cues in addiction: a survey of human neuroimaging studies. Neurosci Biobehav Rev 38:1–16

    Google Scholar 

  • Jeanblanc J, Rolland B, Gierski F, Martinetti MP, Naassila M (2019) Animal models of binge drinking, current challenges to improve face validity. Neurosci Biobehav Rev 106:112–121

    Google Scholar 

  • Jia T, Xie C, Banaschewski T, Barker GJ, Bokde ALW, Buchel C, Quinlan EB, Desrivieres S, Flor H, Grigis A, Garavan H, Gowland P, Heinz A, Ittermann B, Martinot JL, Martinot MP, Nees F, Orfanos DP, Poustka L, Frohner JH, Smolka MN, Walter H, Whelan R, Schumann G, Robbins TW, Feng J, IMAGEN Consortium (2021) Neural network involving medial orbitofrontal cortex and dorsal periaqueductal gray regulation in human alcohol abuse. Sci Adv 7(6)

    Google Scholar 

  • Juarez B, Han MH (2016) Diversity of dopaminergic neural circuits in response to drug exposure. Neuropsychopharmacology 41(10):2424–2446

    Google Scholar 

  • Juarez B, Morel C, Ku SM, Liu Y, Zhang H, Montgomery S, Gregoire H, Ribeiro E, Crumiller M, Roman-Ortiz C, Walsh JJ, Jackson K, Croote DE, Zhu Y, Zhang S, Vendruscolo LF, Edwards S, Roberts A, Hodes GE, Lu Y, Calipari ES, Chaudhury D, Friedman AK, Han MH (2017) Midbrain circuit regulation of individual alcohol drinking behaviors in mice. Nat Commun 8(1):2220

    Google Scholar 

  • June HL, Foster KL, McKay PF, Seyoum R, Woods JE, Harvey SC, Eiler WJ, Grey C, Carroll MR, McCane S, Jones CM, Yin W, Mason D, Cummings R, Garcia M, Ma C, Sarma PV, Cook JM, Skolnick P (2003) The reinforcing properties of alcohol are mediated by GABA(A1) receptors in the ventral pallidum. Neuropsychopharmacology 28(12):2124–2137

    Google Scholar 

  • Jupp B, Krstew E, Dezsi G, Lawrence AJ (2011) Discrete cue-conditioned alcohol-seeking after protracted abstinence: pattern of neural activation and involvement of orexin(1) receptors. Br J Pharmacol 162(4):880–889

    Google Scholar 

  • Kemppainen H, Raivio N, Kiianmaa K (2012) Role for ventral pallidal GABAergic mechanisms in the regulation of ethanol self-administration. Psychopharmacology (Berl) 223(2):211–221

    Google Scholar 

  • Kim CK, Adhikari A, Deisseroth K (2017) Integration of optogenetics with complementary methodologies in systems neuroscience. Nat Rev Neurosci 18(4):222–235

    Google Scholar 

  • Kimbrough A, Lurie DJ, Collazo A, Kreifeldt M, Sidhu H, Macedo GC, D'Esposito M, Contet C, George O (2020) Brain-wide functional architecture remodeling by alcohol dependence and abstinence. Proc Natl Acad Sci U S A 117(4):2149–2159

    Google Scholar 

  • Kircher DM, Aziz HC, Mangieri RA, Morrisett RA (2019) Ethanol experience enhances glutamatergic ventral hippocampal inputs to D1 receptor-expressing medium spiny neurons in the nucleus Accumbens shell. J Neurosci 39(13):2459–2469

    Google Scholar 

  • Koob GF (2008) A role for brain stress systems in addiction. Neuron 59(1):11–34

    Google Scholar 

  • Koob GF, Schulkin J (2019) Addiction and stress: an allostatic view. Neurosci Biobehav Rev 106:245–262

    Google Scholar 

  • Koob GF, Volkow ND (2010) Neurocircuitry of addiction. Neuropsychopharmacology 35(1):217–238

    Google Scholar 

  • Korber C, Sommer WH (2022) From ensembles to meta-ensembles: specific reward encoding by correlated network activity. Front Behav Neurosci 16:977474

    Google Scholar 

  • Kreifeldt M, Herman MA, Sidhu H, Okhuarobo A, Macedo GC, Shahryari R, Gandhi PJ, Roberto M, Contet C (2022) Central amygdala corticotropin-releasing factor neurons promote hyponeophagia but do not control alcohol drinking in mice. Mol Psychiatry 27(5):2502–2513

    Google Scholar 

  • Kupchik YM, Brown RM, Heinsbroek JA, Lobo MK, Schwartz DJ, Kalivas PW (2015) Coding the direct/indirect pathways by D1 and D2 receptors is not valid for accumbens projections. Nat Neurosci 18(9):1230–1232

    Google Scholar 

  • Leriche M, Mendez M, Zimmer L, Berod A (2008) Acute ethanol induces Fos in GABAergic and non-GABAergic forebrain neurons: a double-labeling study in the medial prefrontal cortex and extended amygdala. Neuroscience 153(1):259–267

    Google Scholar 

  • Levine OB, Skelly MJ, Miller JD, Rivera-Irizarry JK, Rowson SA, DiBerto JF, Rinker JA, Thiele TE, Kash TL, Pleil KE (2021) The paraventricular thalamus provides a polysynaptic brake on limbic CRF neurons to sex-dependently blunt binge alcohol drinking and avoidance behavior in mice. Nat Commun 12(1):5080

    Google Scholar 

  • Leyrer-Jackson JM, Hood LE, Olive MF (2021) Alcohol consumption preferentially activates a subset of pro-opiomelanocortin (POMC) producing neurons targeting the amygdala. Neuropharmacology 195:108674

    Google Scholar 

  • Lovelock DF, Tyler RE, Besheer J (2021) Interoception and alcohol: mechanisms, networks, and implications. Neuropharmacology 200:108807

    Google Scholar 

  • Lovinger DM, Alvarez VA (2017) Alcohol and basal ganglia circuitry: animal models. Neuropharmacology 122:46–55

    Google Scholar 

  • Lovinger DM, Roberto M (2023) Synaptic effects induced by alcohol. Curr Top Behav Neurosci

    Google Scholar 

  • Lovinger DM, White G, Weight FF (1989) Ethanol inhibits NMDA-activated ion current in hippocampal neurons. Science 243(4899):1721–1724

    Google Scholar 

  • Luchsinger JR, Fetterly TL, Williford KM, Salimando GJ, Doyle MA, Maldonado J, Simerly RB, Winder DG, Centanni SW (2021) Delineation of an insula-BNST circuit engaged by struggling behavior that regulates avoidance in mice. Nat Commun 12(1):3561

    Google Scholar 

  • Mantsch JR, Baker DA, Funk D, Le AD, Shaham Y (2016) Stress-induced reinstatement of drug seeking: 20 years of progress. Neuropsychopharmacology 41(1):335–356

    Google Scholar 

  • Marinelli PW, Funk D, Juzytsch W, Li Z, LĂŞ AD (2007) Effects of opioid receptor blockade on the renewal of alcohol seeking induced by context: relationship to c-fos mRNA expression. Eur J Neurosci 26(10):2815–2823

    Google Scholar 

  • Marino RAM, Girven KS, Figueiredo A, Navarrete J, Doty C, Sparta DR (2021) Binge ethanol drinking associated with sex-dependent plasticity of neurons in the insula that project to the bed nucleus of the stria terminalis. Neuropharmacology 196:108695

    Google Scholar 

  • Mathis A, Mamidanna P, Cury KM, Abe T, Murthy VN, Mathis MW, Bethge M (2018) DeepLabCut: markerless pose estimation of user-defined body parts with deep learning. Nat Neurosci 21(9):1281–1289

    Google Scholar 

  • Mereu G, Fadda F, Gessa GL (1984) Ethanol stimulates the firing rate of nigral dopaminergic neurons in unanesthetized rats. Brain Res 292(1):63–69

    Google Scholar 

  • Moorman DE (2018) The role of the orbitofrontal cortex in alcohol use, abuse, and dependence. Prog Neuropsychopharmacol Biol Psychiatry 87(Pt A):85–107

    Google Scholar 

  • Morikawa H, Morrisett RA (2010) Ethanol action on dopaminergic neurons in the ventral tegmental area. Functional plasticity and genetic variation: insights into the neurobiology of alcoholism, pp 235–288

    Google Scholar 

  • Munoz B, Fritz BM, Yin F, Atwood BK (2018) Alcohol exposure disrupts mu opioid receptor-mediated long-term depression at insular cortex inputs to dorsolateral striatum. Nat Commun 9(1):1318

    Google Scholar 

  • Naqvi NH, Bechara A (2010) The insula and drug addiction: an interoceptive view of pleasure, urges, and decision-making. Brain Struct Funct 214(5–6):435–450

    Google Scholar 

  • Nestoros JN (1980) Ethanol specifically potentiates GABA-mediated neurotransmission in feline cerebral cortex. Science 209(4457):708–710

    Google Scholar 

  • Nowak KL, McBride WJ, Lumeng L, Li TK, Murphy JM (2000) Involvement of dopamine D2 autoreceptors in the ventral tegmental area on alcohol and saccharin intake of the alcohol-preferring P rat. Alcohol Clin Exp Res 24(4):476–483

    Google Scholar 

  • O'Tousa D, Grahame N (2014) Habit formation: implications for alcoholism research. Alcohol 48(4):327–335

    Google Scholar 

  • Palombo P, Leao RM, Bianchi PC, de Oliveira PEC, Planeta CDS, Cruz FC (2017) Inactivation of the prelimbic cortex impairs the context-induced reinstatement of ethanol seeking. Front Pharmacol 8:725

    Google Scholar 

  • Pati D, Marcinkiewcz CA, DiBerto JF, Cogan ES, McElligott ZA, Kash TL (2020) Chronic intermittent ethanol exposure dysregulates a GABAergic microcircuit in the bed nucleus of the stria terminalis. Neuropharmacology 168:107759

    Google Scholar 

  • Pfarr S, Meinhardt MW, Klee ML, Hansson AC, Vengeliene V, Schonig K, Bartsch D, Hope BT, Spanagel R, Sommer WH (2015) Losing control: excessive alcohol seeking after selective inactivation of Cue-responsive neurons in the infralimbic cortex. J Neurosci 35(30):10750–10761

    Google Scholar 

  • Pfarr S, Schaaf L, Reinert JK, Paul E, Herrmannsdorfer F, Rossmanith M, Kuner T, Hansson AC, Spanagel R, Korber C, Sommer WH (2018) Choice for drug or natural reward engages largely overlapping neuronal ensembles in the infralimbic prefrontal cortex. J Neurosci 38(14):3507–3519

    Google Scholar 

  • Pina MM, Cunningham CL (2017) Ethanol-seeking behavior is expressed directly through an extended amygdala to midbrain neural circuit. Neurobiol Learn Mem 137:83–91

    Google Scholar 

  • Radwanska K, Wrobel E, Korkosz A, Rogowski A, Kostowski W, Bienkowski P, Kaczmarek L (2008) Alcohol relapse induced by discrete cues activates components of AP-1 transcription factor and ERK pathway in the rat basolateral and central amygdala. Neuropsychopharmacology 33(8):1835–1846

    Google Scholar 

  • Rassnick S, Pulvirenti L, Koob GF (1992) Oral ethanol self-administration in rats is reduced by the administration of dopamine and glutamate receptor antagonists into the nucleus accumbens. Psychopharmacology (Berl) 109(1–2):92–98

    Google Scholar 

  • Renteria R, Baltz ET, Gremel CM (2018) Chronic alcohol exposure disrupts top-down control over basal ganglia action selection to produce habits. Nat Commun 9(1):211

    Google Scholar 

  • Renteria R, Cazares C, Baltz ET, Schreiner DC, Yalcinbas EA, Steinkellner T, Hnasko TS, Gremel CM (2021) Mechanism for differential recruitment of orbitostriatal transmission during actions and outcomes following chronic alcohol exposure. eLife 10

    Google Scholar 

  • Rinker JA, Marshall SA, Mazzone CM, Lowery-Gionta EG, Gulati V, Pleil KE, Kash TL, Navarro M, Thiele TE (2017) Extended amygdala to ventral tegmental area corticotropin-releasing factor circuit controls binge ethanol intake. Biol Psychiatry 81(11):930–940

    Google Scholar 

  • Roberto M, Kirson D, Khom S (2021) The role of the central amygdala in alcohol dependence. Cold Spring Harb Perspect Med 11(2)

    Google Scholar 

  • Robins MT, Chiang T, Mores KL, Alongkronrusmee D, van Rijn RM (2018) Critical role for Gi/o-protein activity in the dorsal striatum in the reduction of voluntary alcohol intake in C57Bl/6 mice. Front Psych 9:112

    Google Scholar 

  • Robinson DL, Howard EC, McConnell S, Gonzales RA, Wightman RM (2009) Disparity between tonic and phasic ethanol-induced dopamine increases in the nucleus accumbens of rats. Alcohol Clin Exp Res 33(7):1187–1196

    Google Scholar 

  • Rodd ZA, Melendez RI, Bell RL, Kuc KA, Zhang Y, Murphy JM, McBride WJ (2004) Intracranial self-administration of ethanol within the ventral tegmental area of male Wistar rats: evidence for involvement of dopamine neurons. J Neurosci 24(5):1050–1057

    Google Scholar 

  • Roland AV, Coelho CAO, Haun HL, Gianessi CA, Lopez MF, D'Ambrosio S, Machinski SN, Kroenke CD, Frankland PW, Becker HC, Kash TL (2023) Alcohol dependence modifies brain networks activated during withdrawal and reaccess: a c-fos-based analysis in mice. Biol Psychiatry

    Google Scholar 

  • Roth BL (2016) DREADDs for neuroscientists. Neuron 89(4):683–694

    Google Scholar 

  • Ryabinin AE, Criado JR, Henriksen SJ, Bloom FE, Wilson MC (1997) Differential sensitivity of c-Fos expression in hippocampus and other brain regions to moderate and low doses of alcohol. Mol Psychiatry 2(1):32–43

    Google Scholar 

  • Samson HH, Hodge CW, Tolliver GA, Haraguchi M (1993) Effect of dopamine agonists and antagonists on ethanol-reinforced behavior: the involvement of the nucleus accumbens. Brain Res Bull 30(1–2):133–141

    Google Scholar 

  • Schroeder JP, Spanos M, Stevenson JR, Besheer J, Salling M, Hodge CW (2008) Cue-induced reinstatement of alcohol-seeking behavior is associated with increased ERK1/2 phosphorylation in specific limbic brain regions: blockade by the mGluR5 antagonist MPEP. Neuropharmacology 55(4):546–554

    Google Scholar 

  • Segovia KN, Vontell R, Lopez-Cruz L, Salamone JD, Correa M (2013) c-Fos immunoreactivity in prefrontal, basal ganglia and limbic areas of the rat brain after central and peripheral administration of ethanol and its metabolite acetaldehyde. Front Behav Neurosci 7:48

    Google Scholar 

  • Seif T, Chang SJ, Simms JA, Gibb SL, Dadgar J, Chen BT, Harvey BK, Ron D, Messing RO, Bonci A, Hopf FW (2013) Cortical activation of accumbens hyperpolarization-active NMDARs mediates aversion-resistant alcohol intake. Nat Neurosci 16(8):1094–1100

    Google Scholar 

  • Siciliano CA, Noamany H, Chang CJ, Brown AR, Chen X, Leible D, Lee JJ, Wang J, Vernon AN, Vander Weele CM, Kimchi EY, Heiman M, Tye KM (2019) A cortical-brainstem circuit predicts and governs compulsive alcohol drinking. Science 366(6468):1008–1012

    Google Scholar 

  • Silberman Y, Matthews RT, Winder DG (2013) A corticotropin releasing factor pathway for ethanol regulation of the ventral tegmental area in the bed nucleus of the stria terminalis. J Neurosci 33(3):950–960

    Google Scholar 

  • Sinclair CM, Cleva RM, Hood LE, Olive MF, Gass JT (2012) mGluR5 receptors in the basolateral amygdala and nucleus accumbens regulate cue-induced reinstatement of ethanol-seeking behavior. Pharmacol Biochem Behav 101(3):329–335

    Google Scholar 

  • Smith RJ, Anderson RI, Haun HL, Mulholland PJ, Griffin WC 3rd, Lopez MF, Becker HC (2020) Dynamic c-Fos changes in mouse brain during acute and protracted withdrawal from chronic intermittent ethanol exposure and relapse drinking. Addict Biol 25(6):e12804

    Google Scholar 

  • Thiele TE, Navarro M (2014) "Drinking in the dark" (DID) procedures: a model of binge-like ethanol drinking in non-dependent mice. Alcohol 48(3):235–241

    Google Scholar 

  • Topple AN, Hunt GE, McGregor IS (1998) Possible neural substrates of beer-craving in rats. Neurosci Lett 252(2):99–102

    Google Scholar 

  • Tsurugizawa T, Uematsu A, Uneyama H, Torii K (2010) The role of the GABAergic and dopaminergic systems in the brain response to an intragastric load of alcohol in conscious rats. Neuroscience 171(2):451–460

    Google Scholar 

  • Ueda HR, Dodt HU, Osten P, Economo MN, Chandrashekar J, Keller PJ (2020) Whole-brain profiling of cells and circuits in mammals by tissue clearing and light-sheet microscopy. Neuron 106(3):369–387

    Google Scholar 

  • Vilpoux C, Warnault V, Pierrefiche O, Daoust M, Naassila M (2009) Ethanol-sensitive brain regions in rat and mouse: a cartographic review, using immediate early gene expression. Alcohol Clin Exp Res 33(6):945–969

    Google Scholar 

  • Vranjkovic O, Winkler G, Winder DG (2018) Ketamine administration during a critical period after forced ethanol abstinence inhibits the development of time-dependent affective disturbances. Neuropsychopharmacology 43(9):1915–1923

    Google Scholar 

  • Wandres M, Pfarr S, Molnar B, Schollkopf U, Ercsey-Ravasz M, Sommer WH, Korber C (2021) Alcohol and sweet reward are encoded by distinct meta-ensembles. Neuropharmacology 195:108496

    Google Scholar 

  • Wedzony K, Koros E, Czyrak A, Chocyk A, Czepiel K, Fijal K, Mackowiak M, Rogowski A, Kostowski W, Bienkowski P (2003) Different pattern of brain c-Fos expression following re-exposure to ethanol or sucrose self-administration environment. Naunyn Schmiedebergs Arch Pharmacol 368(5):331–341

    Google Scholar 

  • Weiss F, Lorang MT, Bloom FE, Koob GF (1993) Oral alcohol self-administration stimulates dopamine release in the rat nucleus accumbens: genetic and motivational determinants. J Pharmacol Exp Ther 267(1):250–258

    Google Scholar 

  • Wills TA, Kash TL, Winder DG (2013) Developmental changes in the acute ethanol sensitivity of glutamatergic and GABAergic transmission in the BNST. Alcohol 47(7):531–537

    Google Scholar 

  • Witkiewitz K, Stein ER, Votaw VR, Hallgren KA, Gibson BC, Boness CL, Pearson MR, Maisto SA (2022) Constructs derived from the addiction cycle predict alcohol use disorder treatment outcomes and recovery 3 years following treatment. Psychol Addict Behav

    Google Scholar 

  • You C, Vandegrift B, Brodie MS (2018) Ethanol actions on the ventral tegmental area: novel potential targets on reward pathway neurons. Psychopharmacology (Berl) 235(6):1711–1726

    Google Scholar 

  • Zhao Y, Dayas CV, Aujla H, Baptista MA, Martin-Fardon R, Weiss F (2006) Activation of group II metabotropic glutamate receptors attenuates both stress and cue-induced ethanol-seeking and modulates c-fos expression in the hippocampus and amygdala. J Neurosci 26(39):9967–9974

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Danny G. Winder .

Rights and permissions

Reprints and permissions

Copyright information

© 2023 The Author(s), under exclusive license to Springer Nature Switzerland AG

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Doyle, M.A., Taylor, A., Winder, D.G. (2023). Neural Circuitries and Alcohol Use Disorder: Cutting Corners in the Cycle. In: Current Topics in Behavioral Neurosciences. Springer, Berlin, Heidelberg. https://doi.org/10.1007/7854_2023_454

Download citation

  • DOI: https://doi.org/10.1007/7854_2023_454

  • Published:

  • Publisher Name: Springer, Berlin, Heidelberg

Publish with us

Policies and ethics