We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Published Online:https://doi.org/10.2217/fnl.10.14

The progression from recreational drug use to drug addiction impacts multiple neurobiological processes and can be conceptualized as a transition from positive to negative reinforcement mechanisms driving both drug-taking and drug-seeking behaviors. Neurobiological mechanisms for negative reinforcement, defined as drug taking that alleviates a negative emotional state, involve changes in the brain reward system and recruitment of brain stress (or antireward) systems within forebrain structures, including the extended amygdala. These systems are hypothesized to be dysregulated by excessive drug intake and to contribute to allostatic changes in reinforcement mechanisms associated with addiction. Points of intersection between positive and negative motivational circuitry may further drive the compulsivity of drug addiction but also provide a rich neurobiological substrate for therapeutic intervention.

Bibliography

Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

References

  • American Psychiatric Association: Diagnositc and Statistical Manual of Mental Disorders (4th Edition). American Psychiatric Press, Washington, DC, USA (2000).
  • Koob GF, Le Moal M: Drug abuse: hedonic homeostatic dysregulation. Science278,52–58 (1997).▪▪ Theoretical review arguing that drug addiction results from a dysregulation of reward homeostasis.
  • Grant BF, Dawson DA: Age of onset of drug use and its association with DSM-IV drug abuse and dependence: results from the National Longitudinal Alcohol Epidemiologic Survey. J. Subst. Abuse10,163–173 (1998).
  • Grant BF, Dawson DA, Stinson FS, Chou SP, Dufour MC, Pickering RP: The 12-month prevalence and trends in DSM-IV alcohol abuse and dependence: United States, 1991–1992 and 2001–2002. Drug Alcohol Depend.74,223–234 (2004).
  • Substance Abuse and Mental Health Services Administration: Results from the 2007 National Survey on Drug Use and Health: National Findings. Office of Applied Statistics, MD, USA (2008).
  • Bindra D: A Theory of Intelligent Behavior. Wiley, NY, USA (1976).
  • Robinson TE, Berridge KC: The neural basis of drug craving: an incentive-sensitization theory of addiction, Brain Res. Rev.18,247–291 (1993).
  • Hyman SE, Malenka RC, Nestler EJ: Neural mechanisms of addiction: the role of reward-related learning and memory. Annu. Rev. Neurosci.29,565–598 (2006).▪ Hypothesizes that the persistence of drug addiction results from a perturbation of reward-related learning and memory processes.
  • Kalivas PW, Volkow ND: The neural basis of addiction: a pathology of motivation and choice. Am. J. Psychiatry162,1403–1413 (2005).
  • 10  Solomon RL, Corbit JD: An opponent-process theory of motivation: 1. Temporal dynamics of affect. Psychol. Rev.81,119–145 (1974).
  • 11  Siegel S: Evidence from rats that morphine tolerance is a learned response. J. Comp. Physiol. Psychol.89,498–506 (1975).
  • 12  Poulos CX, Cappell H: Homeostatic theory of drug tolerance: a general model of physiological adaptation. Psychol. Rev.98,390–408 (1991).
  • 13  Koob GF, Le Moal M: Drug addiction, dysregulation of reward, and allostasis. Neuropsychopharmacology24,97–129 (2001).
  • 14  Koob GF, Le Moal M: Addiction and the brain antireward system. Annu. Rev. Psychol.59,29–53 (2008).
  • 15  Koob GF, Bloom FE: Cellular and molecular mechanisms of drug dependence. Science242,715–723 (1988).
  • 16  Koob GF: Neuroadaptive mechanisms of addiction: studies on the extended amygdala. Eur. Neuropsychopharmacol.13,442–452 (2003).
  • 17  Nestler EJ: Molecular basis of long-term plasticity underlying addiction. Nat. Rev. Neurosci.2,119–128 (2001).
  • 18  Aston-Jones G, Delfs JM, Druhan J, Zhu Y: The bed nucleus of the stria terminalis: a target site for noradrenergic actions in opiate withdrawal. In: Advancing from the Ventral Striatum to the Extended Amygdala: Implications for Neuropsychiatry and Drug Abuse. McGinty JF (Ed.). New York Academy of Sciences, NY, USA, 486–498 (1999).
  • 19  Sterling P, Eyer J: Allostasis: a new paradigm to explain arousal pathology. In: Handbook of Life Stress, Cognition and Health. Fisher S, Reason J (Eds). John Wiley and Sons, UK, 629–649 (1988).
  • 20  McEwen BS: Allostasis and allostatic load: implications for neuropsychopharmacology. Neuropsychopharmacology22,108–124 (2000).
  • 21  Olds J, Milner P: Positive reinforcement produced by electrical stimulation of septal area and other regions of rat brain. J. Comp. Physiol. Psychol.47,419–427 (1954).
  • 22  Koob GF, Winger GD, Meyerhoff JL, Annau Z: Effects of D-amphetamine on concurrent self-stimulation of forebrain and brain stem loci. Brain Res.137,109–126 (1977).
  • 23  Simon H, Stinus L, Tassin JP et al.: Is the dopaminergic mesocorticolimbic system necessary for intracranial self-stimulation? Biochemical and behavioral studies from A10 cell bodies and terminals. Behav. Neural Biol.27,125–145 (1979).
  • 24  Kornetsky C, Esposito RU: Euphorigenic drugs: effects on the reward pathways of the brain. Fed. Proc.38,2473–2476 (1979).
  • 25  Hernandez G, Hamdani S, Rajabi H et al.: Prolonged rewarding stimulation of the rat medial forebrain bundle: neurochemical and behavioral consequences. Behav. Neurosci.120,888–904 (2006).
  • 26  Garris PA, Kilpatrick M, Bunin MA, Michael D, Walker QD, Wightman RM: Dissociation of dopamine release in the nucleus accumbens from intracranial self-stimulation. Nature398,67–69 (1999).
  • 27  Miliaressis E, Emond C, Merali Z: Re-evaluation of the role of dopamine in intracranial self-stimulation using in vivo microdialysis. Behav. Brain Res.46,43–48 (1991).
  • 28  Robbins TW: Relationship between reward-enhancing and stereotypical effects of psychomotor stimulant drugs. Nature264,57–59 (1976).
  • 29  Miliaressis E, Le Moal M: Stimulation of the medial forebrain bundle: behavioral dissociation of its rewarding and activating effects. Neurosci. Lett.2,295–300 (1976).
  • 30  Salamone JD, Correa M, Farrar A, Mingote SM: Effort-related functions of nucleus accumbens dopamine and associated forebrain circuits. Psychopharmacology191,461–482 (2007).
  • 31  Le Moal M, Simon H: Mesocorticolimbic dopaminergic network: functional and regulatory roles. Physiol. Rev.71,155–234 (1991).
  • 32  Robbins TW, Everitt BJ: A role for mesencephalic dopamine in activation: commentary on Berridge (2006). Psychopharmacology (Berl.)191,433–437 (2007).
  • 33  Shippenberg TS, Koob GF: Recent advances in animal models of drug addiction and alcoholism. In: Neuropsychopharmacology: The Fifth Generation of Progress. Davis KL, Charney D, Coyle JT, Nemeroff C (Eds). Lippincott Williams and Wilkins, PA, USA, in vivo1381–1397 (2002).
  • 34  Koob GF: The neurobiology of addiction: a neuroadaptational view relevant for diagnosis. Addiction101(Suppl. 1),23–30 (2006).
  • 35  Nestler EJ: Is there a common molecular pathway for addiction? Nat. Neurosci.8,1445–1449 (2005).
  • 36  Sarnyai Z, Biro E, Gardi J, Vecsernyes M, Julesz J, Telegdy G: Brain corticotropin-releasing factor mediates ‘anxiety-like’ behavior induced by cocaine withdrawal in rats. Brain Res.675,89–97 (1995).
  • 37  Basso AM, Spina M, Rivier J, Vale W, Koob GF: Corticotropin-releasing factor antagonist attenuates the ‘anxiogenic-like’ effect in the defensive burying paradigm but not in the elevated plus-maze following chronic cocaine in rats. Psychopharmacology (Berl.)145,21–30 (1999).
  • 38  Schulteis G, Stinus L, Risbrough VB, Koob GF: Clonidine blocks acquisition but not expression of conditioned opiate withdrawal in rats. Neuropsychopharmacology19,406–416 (1998).
  • 39  Harris GC, Aston-Jones G: β-Adrenergic antagonists attenuate withdrawal anxiety in cocaine- and morphine-dependent rats. Psychopharmacology (Berl.)113,131–136 (1993).
  • 40  Stinus L, Le Moal M, Koob GF: Nucleus accumbens and amygdala are possible substrates for the aversive stimulus effects of opiate withdrawal. Neuroscience37,767–773 (1990).
  • 41  Charlton BG, Ferrier IN, Perry RH: Distribution of corticotropin-releasing factor-like immunoreactivity in human brain. Neuropeptides10,329–334 (1987).
  • 42  Swanson LW, Sawchenko PE, Rivier J, Vale W: The organization of ovine corticotropin-releasing factor immunoreactive cells and fibers in the rat brain: an immunohistochemical study. Neuroendocrinology36,165–186 (1983).
  • 43  Stinus L, Cador M, Zorrilla EP, Koob GF: Buprenorphine and a CRF1 antagonist block the acquisition of opiate withdrawal-induced conditioned place aversion in rats. Neuropsychopharmacology30,90–98 (2005).
  • 44  Heinrichs SC, Menzaghi F, Schulteis G, Koob GF, Stinus L: Suppression of corticotropin-releasing factor in the amygdala attenuates aversive consequences of morphine withdrawal. Behav. Pharmacol.6,74–80 (1995).
  • 45  Baldwin HA, Rassnick S, Rivier J, Koob GF, Britton KT: CRF antagonist reverses the ‘anxiogenic’ response to ethanol withdrawal in the rat. Psychopharmacology (Berl.)103,227–232 (1991).
  • 46  Knapp DJ, Overstreet DH, Moy SS, Breese GR: SB242084, flumazenil, and CRA1000 block ethanol withdrawal-induced anxiety in rats. Alcohol32,101–111 (2004).
  • 47  Overstreet DH, Knapp DJ, Breese GR: Modulation of multiple ethanol withdrawal-induced anxiety-like behavior by CRF and CRF1 receptors. Pharmacol. Biochem. Behav.77,405–413 (2004).
  • 48  Funk CK, Zorrilla EP, Lee M-J, Rice KC, Koob GF: Corticotropin-releasing factor 1 antagonists selectively reduce ethanol self-administration in ethanol-dependent rats. Biol. Psychiatry61,78–86 (2007).
  • 49  Tucci S, Cheeta S, Seth P, File SE: Corticotropin releasing factor antagonist, a-helical CRF9–41, reverses nicotine-induced conditioned, but not unconditioned, anxiety, Psychopharmacology (Berl.)167,251–256 (2003).
  • 50  George O, Ghozland S, Azar MR et al.: CRF-CRF1 system activation mediates withdrawal-induced increases in nicotine self-administration in nicotine-dependent rats. Proc. Natl Acad. Sci. USA104,17198–17203 (2007).
  • 51  Rassnick S, Heinrichs SC, Britton KT, Koob GF: Microinjection of a corticotropin-releasing factor antagonist into the central nucleus of the amygdala reverses anxiogenic-like effects of ethanol withdrawal. Brain Res.605,25–32 (1993).
  • 52  Breese GR, Overstreet DH, Knapp DJ, Navarro M: Prior multiple ethanol withdrawals enhance stress-induced anxiety-like behavior: inhibition by CRF1- and benzodiazepine-receptor antagonists and a 5-HT1a-receptor agonist. Neuropsychopharmacology30,1662–1669 (2005).
  • 53  Valdez GR, Zorrilla EP, Roberts AJ, Koob GF: Antagonism of corticotropin-releasing factor attenuates the enhanced responsiveness to stress observed during protracted ethanol abstinence. Alcohol29,55–60 (2003).
  • 54  Koob GF: Neurobiological substrates for the dark side of compulsivity in addiction. Neuropharmacology56(Suppl. 1),18–31 (2009).
  • 55  Richter RM, Weiss F: In vivo CRF release in rat amygdala is increased during cocaine withdrawal in self-administering rats. Synapse32,254–261 (1999).
  • 56  Weiss F, Ciccocioppo R, Parsons LH et al.: Compulsive drug-seeking behavior and relapse: neuroadaptation, stress, and conditioning factors. In: QuinonesThe Biological Basis of Cocaine Addiction. Jenab V (Ed.). New York Academy of Sciences, NY, USA, 1–26 (2001).
  • 57  Matta SG, Valentine JD, Sharp BM: Nicotinic activation of CRH neurons in extrahypothalamic regions of the rat brain. Endocrine7,245–253 (1997).
  • 58  Slawecki CJ, Thorsell AK, Khoury AE, Mathe AA, Ehlers CL: Increased CRF-like and NPY-like immunoreactivity in adult rats exposed to nicotine during adolescence: relation to anxiety-like and depressive-like behavior. Neuropeptides39,369–377 (2005).
  • 59  Funk CK, O’Dell LE, Crawford EF, Koob GF: Corticotropin-releasing factor within the central nucleus of the amygdala mediates enhanced ethanol self-administration in withdrawn, ethanol-dependent rats. J. Neurosci.26,11324–11332 (2006).▪ Demonstrates that blockade of corticotropin-releasing factor receptors specifically within the central nucleus of the amygdala reduces the increased alcohol drinking associated with dependence without altering alcohol intake in nondependent animals.
  • 60  Merlo-Pich E, Lorang M, Yeganeh M et al.: Increase of extracellular corticotropin-releasing factor-like immunoreactivity levels in the amygdala of awake rats during restraint stress and ethanol withdrawal as measured by microdialysis. J. Neurosci.15,5439–5447 (1995).
  • 61  Olive MF, Koenig HN, Nannini MA, Hodge CW: Elevated extracellular CRF levels in the bed nucleus of the stria terminalis during ethanol withdrawal and reduction by subsequent ethanol intake. Pharmacol. Biochem. Behav.72,213–220 (2002).
  • 62  Rodriguez de Fonseca F, Carrera MRA, Navarro M, Koob GF, Weiss F: Activation of corticotropin-releasing factor in the limbic system during cannabinoid withdrawal. Science276,2050–2054 (1997).
  • 63  Specio SE, Wee S, O’Dell LE, Boutrel B, Zorrilla EP, Koob GF: CRF1 receptor antagonists attenuate escalated cocaine self-administration in rats. Psychopharmacology (Berl.)196,473–482 (2008).
  • 64  Greenwell TN, Funk CK, Cottone P et al.: Corticotropin-releasing factor-1 receptor antagonists decrease heroin self-administration in long-, but not short-access rats. Addict. Biol.14,130–143 (2009).
  • 65  Valdez GR, Sabino V, Koob GF: Increased anxiety-like behavior and ethanol self-administration in dependent rats: reversal via corticotropin-releasing factor-2 receptor activation. Alcohol. Clin. Exp. Res.28,865–872 (2004).
  • 66  Rimondini R, Arlinde C, Sommer W, Heilig M: Long-lasting increase in voluntary ethanol consumption and transcriptional regulation in the rat brain after intermittent exposure to alcohol. FASEB J.16,27–35 (2002).
  • 67  Treutlein J, Kissling C, Frank J et al.: Genetic association of the human corticotropin releasing hormone receptor 1 (CRHR1) with binge drinking and alcohol intake patterns in two independent samples. Mol. Psychiatry11,594–602 (2006).
  • 68  Blomeyer D, Treutlein J, Esser G et al.: Interaction between CRHR1 gene and stressful life events predicts adolescent heavy alcohol use. Biol. Psychiatry63,146–151 (2008).
  • 69  Morilak DA, Barrera G, Echevarria DJ et al.: Role of brain norepinephrine in the behavioral response to stress. Prog. Neuropsychopharmacol. Biol. Psychiatry29,1214–1224 (2005).
  • 70  Maynert EW, Levi R: Stress-induced release of brain norepinephrine and its inhibition by drugs. J. Pharmacol. Exp. Ther.143,90–95 (1964).
  • 71  Aston-Jones G, Cohen JD: An integrative theory of locus coeruleus-norepinephrine function: adaptive gain and optimal performance. Annu. Rev. Neurosci.28,403–450 (2005).
  • 72  Koob GF: Corticotropin-releasing factor, norepinephrine and stress. Biol. Psychiatry46,1167–1180 (1999).
  • 73  Delfs JM, Zhu Y, Druhan JP, Aston-Jones G: Noradrenaline in the ventral forebrain is critical for opiate withdrawal-induced aversion. Nature403,430–434 (2000).
  • 74  Nestler EJ, Alreja M, Aghajanian GK: Molecular and cellular mechanisms of opiate action: studies in the rat locus coeruleus. Brain Res. Bull.35,521–528 (1994).
  • 75  Borg S, Czamecka A, Kvande H, Mossberg D, Sedvall G: Clinical conditions and concentrations of MOPEG in the cerebrospinal fluid of male alcoholic patients during withdrawal. Alcohol. Clin. Exp. Res.9,103–108 (1985).
  • 76  Borg S, Kvande H, Sedvall G: Central norepinephrine metabolism during alcohol intoxication in addicts and healthy volunteers. Science213,1135–1137 (1981).
  • 77  Fujimoto A, Nagao T, Ebara T, Sato M, Otsuki S: Cerebrospinal fluid monoamine metabolites during alcohol withdrawal syndrome and recovered state. Biol. Psychiatry18,1141–1152 (1983).
  • 78  Amit Z, Brown ZW, Levitan DE, Ogren SO: Noradrenergic mediation of the positive reinforcing properties of ethanol: I. Suppression of ethanol consumption in laboratory rats following dopamine-β-hydroxylase inhibition. Arch. Int. Pharmacodyn. Ther.230,65–75 (1977).
  • 79  Brown ZW, Amit Z: The effects of selective catecholamine depletions by 6-hydroxydopamine on ethanol preference in rats. Neurosci. Lett.5,333–336 (1977).
  • 80  Davis WM, Werner TE, Smith SG: Reinforcement with intragastric infusions of ethanol: blocking effect of FLA 57. Pharmacol. Biochem. Behav.11,545–548 (1979).
  • 81  Ventura R, De Carolis D, Alcaro A, Puglisi-Allegra S: Ethanol consumption and reward depend on norepinephrine in the prefrontal cortex. Neuroreport17,1813–1817 (2006).
  • 82  Weinshenker D, Rust NC, Miller NS, Palmiter RD: Ethanol-associated behaviors of mice lacking norepinephrine. J. Neurosci.20,3157–3164 (2000).
  • 83  Walker BM, Rasmussen DD, Raskind MA, Koob GF: α1-noradrenergic receptor antagonism blocks dependence-induced increases in responding for ethanol. Alcohol42,91–97 (2008).
  • 84  Wee S, Mandyam CD, Lekic DM, Koob GF: α1-noradrenergic system role in increased motivation for cocaine intake in rats with prolonged access. Eur. Neuropsychopharmacol.18,303–311 (2008).
  • 85  Greenwell TN, Walker BM, Cottone P, Zorrilla EP, Koob GF: The α1 adrenergic receptor antagonist prazosin reduces heroin self-administration in rats with extended access to heroin administration. Pharmacol. Biochem. Behav.91,295–302 (2009).
  • 86  Valentino RJ, Foote SL, Page ME: The locus coeruleus as a site for integrating corticotropin-releasing factor and noradrenergic mediation of stress responses. In: Corticotropin-Releasing Factor and Cytokines: Role in the Stress Response. Tache Y, Rivier C (Eds). New York Academy of Sciences, NY, USA, 173–188 (1993).
  • 87  Valentino RJ, Page ME, Curtis AL: Activation of noradrenergic locus coeruleus neurons by hemodynamic stress is due to local release of corticotropin-releasing factor. Brain Res.555,25–34 (1991).
  • 88  Van Bockstaele EJ, Colago EE, Valentino RJ: Amygdaloid corticotropin-releasing factor targets locus coeruleus dendrites: substrate for the co-ordination of emotional and cognitive limbs of the stress response. J. Neuroendocrinol.10,743–757 (1998).
  • 89  Alonso G, Szafarczyk A, Balmefrezol M, Assenmacher I: Immunocytochemical evidence for stimulatory control by the ventral noradrenergic bundle of parvocellular neurons of the paraventricular nucleus secreting corticotropin-releasing hormone and vasopressin in rats. Brain Res.397,297–307 (1986).
  • 90  Chavkin C, James IF, Goldstein A: Dynorphin is a specific endogenous ligand of the κ opioid receptor. Science215,413–415 (1982).
  • 91  Watson SJ, Khachaturian H, Akil H, Coy DH, Goldstein A: Comparison of the distribution of dynorphin systems and enkephalin systems in brain. Science218,1134–1136 (1982).
  • 92  Fallon JH, Leslie FM: Distribution of dynorphin and enkephalin peptides in the rat brain. J. Comp. Neurol.249,293–336 (1986).
  • 93  Mucha RF, Herz A: Motivational properties of κ and µ opioid receptor agonists studied with place and taste preference conditioning. Psychopharmacology (Berl.)86,274–280 (1985).
  • 94  Pfeiffer A, Brantl V, Herz A, Emrich HM: Psychotomimesis mediated by κ opiate receptors. Science233,774–776 (1986).
  • 95  Todtenkopf MS, Marcus JF, Portoghese PS, Carlezon WA Jr: Effects of κ-opioid receptor ligands on intracranial self-stimulation in rats. Psychopharmacology (Berl.)172,463–470 (2004).
  • 96  Spanagel R, Herz A, Shippenberg TS: Opposing tonically active endogenous opioid systems modulate the mesolimbic dopaminergic pathway. Proc. Natl Acad. Sci. USA89,2046–2050 (1992).
  • 97  Di Chiara G, Imperato A: Drugs abused by humans preferentially increase synaptic dopamine concentrations in the mesolimbic system of freely moving rats. Proc. Natl Acad. Sci. USA85,5274–5278 (1988).
  • 98  Margolis EB, Hjelmstad GO, Bonci A, Fields HL: κ-opioid agonists directly inhibit midbrain dopaminergic neurons. J. Neurosci.23,9981–9986 (2003).
  • 99  Sivam SP: Cocaine selectively increases striatonigral dynorphin levels by a dopaminergic mechanism. J. Pharmacol. Exp. Ther.250,818–824 (1989).
  • 100  Spangler R, Unterwald EM, Kreek MJ: ‘Binge’ cocaine administration induces a sustained increase of prodynorphin mRNA in rat caudate-putamen. Mol. Brain Res.19,323–327 (1993).
  • 101  Daunais JB, Roberts DC, McGinty JF: Cocaine self-administration increases preprodynorphin, but not c-fos, mRNA in rat striatum. Neuroreport4,543–546 (1993).
  • 102  Unterwald EM, Rubenfeld JM, Kreek MJ: Repeated cocaine administration upregulates κ and µ, but not δ, opioid receptors. Neuroreport5,1613–1616 (1994).
  • 103  Rosin A, Lindholm S, Franck J, Georgieva J: Downregulation of κ opioid receptor mRNA levels by chronic ethanol and repetitive cocaine in rat ventral tegmentum and nucleus accumbens. Neurosci. Lett.275,1–4 (1999).
  • 104  Lindholm S, Ploj K, Franck J, Nylander I: Repeated ethanol administration induces short- and long-term changes in enkephalin and dynorphin tissue concentrations in rat brain. Alcohol22,165–171 (2000).
  • 105  Rattan AK, Koo KL, Tejwani GA, Bhargava HN: The effect of morphine tolerance dependence and abstinence on immunoreactive dynorphin (1–13) levels in discrete brain regions, spinal cord, pituitary gland and peripheral tissues of the rat. Brain Res.584,207–212 (1992).
  • 106  Turchan J, Lason W, Budziszewska B, Przewlocka B: Effects of single and repeated morphine administration on the prodynorphin, proenkephalin and dopamine D2 receptor gene expression in the mouse brain. Neuropeptides31,24–28 (1997).
  • 107  Walker BM, Koob GF: Pharmacological evidence for a motivational role of κ-opioid systems in ethanol dependence. Neuropsychopharmacology33,643–652 (2008).
  • 108  Doyon WM, Howard EC, Shippenberg TS, Gonzales RA: κ-opioid receptor modulation of accumbal dopamine concentration during operant ethanol self-administration. Neuropharmacology51,487–496 (2006).
  • 109  Kovacs KM, Szakall I, O’Brien D et al.: Decreased oral self-administration of alcohol in κ-opioid receptor knock-out mice. Alcohol. Clin. Exp. Res.29,730–738 (2005).
  • 110  Negus SS: Effects of the κ opioid agonist U50,488 and the κ opioid antagonist nor-binaltorphimine on choice between cocaine and food in rhesus monkeys. Psychopharmacology176,204–213 (2004).
  • 111  Negus SS, Henriksen SJ, Mattox A et al.: Effect of antagonists selective for µ, δ and κ opioid receptors on the reinforcing effects of heroin in rats. J. Pharmacol. Exp. Ther.265,1245–1252 (1993).
  • 112  Wee S, Orio L, Ghirmai S, Cashman J, Koob GF: Inhibition of κ opioid receptors attenuates the increased motivation for cocaine in rats with extended access to cocaine. Psychopharmacology (Berl.)205,565–575 (2009).
  • 113  Xi ZX, Fuller SA, Stein EA: Dopamine release in the nucleus accumbens during heroin self-administration is modulated by κ opioid receptors: an in vivo fast-cyclic voltammetry study. J. Pharmacol. Exp. Ther.284,151–161 (1998).
  • 114  Shirayama Y, Ishida H, Iwata M, Hazama GI, Kawahara R, Duman RS: Stress increases dynorphin immunoreactivity in limbic brain regions and dynorphin antagonism produces antidepressant-like effects. J. Neurochem.90,1258–1268 (2004).
  • 115  Land BB, Bruchas MR, Lemos JC, Xu M, Melief EJ, Chavkin C: The dysphoric component of stress is encoded by activation of the dynorphin κ-opioid system. J. Neurosci.28,407–414 (2008).
  • 116  McLaughlin JP, Marton-Popovici M, Chavkin C: κ opioid receptor antagonism and prodynorphin gene disruption block stress-induced behavioral responses. J. Neurosci.23,5674–5683 (2003).
  • 117  Beardsley PM, Howard JL, Shelton KL, Carroll FI: Differential effects of the novel κ opioid receptor antagonist, JDTic, on reinstatement of cocaine-seeking induced by footshock stressors vs cocaine primes and its antidepressant-like effects in rats. Psychopharmacology (Berl.)183,118–126 (2005).
  • 118  Valdez GR, Platt DM, Rowlett JK, Rüedi-Bettschen D, Spealman RD: κ agonist-induced reinstatement of cocaine seeking in squirrel monkeys: a role for opioid and stress-related mechanisms. J. Pharmacol. Exp. Ther.323,525–533 (2007).
  • 119  Zimprich A, Kraus J, Woltje M et al.: An allelic variation in the human prodynorphin gene promoter alters stimulus-induced expression. J. Neurochem.74,472–477 (2000).
  • 120  Nomura A, Ujike H, Tanaka Y et al.: Genetic variant of prodynorphin gene is risk factor for methamphetamine dependence. Neurosci. Lett.400,158–162 (2006).
  • 121  Ray R, Doyle GA, Crowley JJ et al.: A functional prodynorphin promoter polymorphism and opioid dependence. Psychiatr. Genet.15,295–298 (2005).
  • 122  Williams TJ, LaForge KS, Gordon D et al.: Prodynorphin gene promoter repeat associated with cocaine/alcohol codependence. Addict. Biol.12,496–502 (2007).
  • 123  Yuferov V, Ji F, Nielsen DA et al.: A functional haplotype implicated in vulnerability to develop cocaine dependence is associated with reduced PDYN expression in human brain. Neuropsychopharmacology34,1185–1197 (2009).
  • 124  Kalivas PW: How do we determine which drug-induced neuroplastic changes are important? Nat. Neurosci.8,1440–1441 (2005).▪ Argues for the importance of meticulously studying the precise functions of drug-induced neuroadaptations in animal models across the addiction timeline.
  • 125  Rossetti ZL, Hmaidan Y, Gessa GL: Marked inhibition of mesolimbic dopamine release: a common feature of ethanol, morphine, cocaine and amphetamine abstinence in rats. Eur. J. Pharmacol.221,227–234 (1992).
  • 126  Weiss F, Markou A, Lorang MT, Koob GF: Basal extracellular dopamine levels in the nucleus accumbens are decreased during cocaine withdrawal after unlimited-access self-administration. Brain Res.593,314–318 (1992).
  • 127  Weiss F, Parsons LH, Schulteis G et al.: Ethanol self-administration restores withdrawal-associated deficiencies in accumbal dopamine and 5-hydroxytryptamine release in dependent rats. J. Neurosci.16,3474–3485 (1996).
  • 128  Melis M, Spiga S, Diana M: The dopamine hypothesis of drug addiction: hypodopaminergic state. Int. Rev. Neurobiol.63,101–154 (2005).
  • 129  Markou A, Koob GF: Bromocriptine reverses the elevation in intracranial self-stimulation thresholds observed in a rat model of cocaine withdrawal. Neuropsychopharmacology7,213–224 (1992).
  • 130  Chartoff EH, Mague SD, Barhight MF, Smith AM, Carlezon WA Jr: Behavioral and molecular effects of dopamine D1 receptor stimulation during naloxone-precipitated morphine withdrawal. J. Neurosci.26,6450–6457 (2006).
  • 131  Liu ZH, Shin R, Ikemoto S: Dual role of medial A10 dopamine neurons in affective encoding. Neuropsychopharmacology33,3010–3020 (2008).
  • 132  Alheid GF, Heimer L: New perspectives in basal forebrain organization of special relevance for neuropsychiatric disorders: the striatopallidal, amygdaloid, and corticopetal components of substantia innominata. Neuroscience27,1–39 (1988).
  • 133  Johnston JB: Further contributions to the study of the evolution of the forebrain. J. Comp. Neurol.35,337–481 (1923).
  • 134  Heimer L, Alheid G: Piecing together the puzzle of basal forebrain anatomy. In: The Basal Forebrain: Anatomy to Function. Napier TC, Kalivas PW, Hanin I (Eds). Plenum Press, NY, USA, 1–42 (1991).
  • 135  Heimer L, Zahm DS, Churchill L, Kalivas PW, Wohltmann C: Specificity in the projection patterns of accumbal core and shell in the rat. Neuroscience41,89–125 (1991).
  • 136  Koob GF, Le Moal M: Plasticity of reward neurocircuitry and the ‘dark side’ of drug addiction. Nat. Neurosci.8,1442–1444 (2005).
  • 137  Terwilliger RZ, Beitner-Johnson D, Sevarino KA, Crain SM, Nestler EJ: A general role for adaptations in G-proteins and the cyclic AMP system in mediating the chronic actions of morphine and cocaine on neuronal function. Brain Res.548,100–110 (1991).
  • 138  Striplin CD, Kalivas PW: Robustness of G protein changes in cocaine sensitization shown with immunoblotting. Synapse14,10–15 (1993).
  • 139  Nestler EJ, Terwilliger RZ, Walker JR, Sevarino KA, Duman RS: Chronic cocaine treatment decreases levels of the G protein subunits Gi α and Go α in discrete regions of rat brain. J. Neurochem.55,1079–1082 (1990).
  • 140  McLeman ER, Warsh JJ, Ang L et al.: The human nucleus accumbens is highly susceptible to G protein down-regulation by methamphetamine and heroin. J. Neurochem.74,2120–2126 (2000).
  • 141  Ortiz J, Fitzgerald LW, Lane S, Terwilliger R, Nestler EJ: Biochemical adaptations in the mesolimbic dopamine system in response to repeated stress. Neuropsychopharmacology14,443–452 (1996).
  • 142  Self DW, Terwilliger RZ, Nestler EJ, Stein L: Inactivation of Gi and Go proteins in nucleus accumbens reduces both cocaine and heroin reinforcement. J. Neurosci.14,6239–6247 (1994).
  • 143  Ahmed SH, Koob GF: Transition from moderate to excessive drug intake: change in hedonic set point. Science282,298–300 (1998).
  • 144  Edwards S, Graham DL, Bachtell RK, Self DW: Region-specific tolerance to cocaine-regulated cAMP-dependent protein phosphorylation following chronic self-administration. Eur. J. Neurosci.25,2201–2213 (2007).
  • 145  Edwards S, Graham DL, Whisler KN, Self DW: Phosphorylation of GluR1, ERK, and CREB during spontaneous withdrawal from chronic heroin self-administration. Synapse63,224–235 (2009).
  • 146  Self DW, Genova LM, Hope BT, Barnhart WJ, Spencer JJ, Nestler EJ: Involvement of cAMP-dependent protein kinase in the nucleus accumbens in cocaine self-administration and relapse of cocaine-seeking behavior J. Neurosci.18,1848–1859 (1998).
  • 147  Whisler K, Hall S, Edwards S, Akkentli F, Self DW: Tonic activation of NAc PKA signaling facilitates D2 receptor-mediated relapse to cocaine seeking and locomotor activity. Abstr. Soc. Neurosci.29,420.417 (2003).
  • 148  Self DW: Regulation of drug-taking and -seeking behaviors by neuroadaptations in the mesolimbic dopamine system. Neuropharmacology47(Suppl. 1),242–255 (2004).
  • 149  Nestler EJ: Historical review: molecular and cellular mechanisms of opiate and cocaine addiction. Trends Pharmacol. Sci.25,210–218 (2004).
  • 150  Larson EB, Whisler K, Smagula C et al.: Preferential over-expression of Gs-α in direct and indirect striatal output pathways differentially alters cocaine-taking and -seeking behaviors. Soc. Neurosci. Abstr.35 (2009) (Abstract 649.11).
  • 151  Sorg BA, Kalivas PW: Effects of cocaine and footshock stress on extracellular dopamine levels in the ventral striatum. Brain Res.559,29–36 (1991).
  • 152  Pan JT, Lookingland KJ, Moore KE: Differential effects of corticotropin-releasing hormone on central dopaminergic and noradrenergic neurons. J. Biomed. Sci.2,50–56 (1995).
  • 153  Lodge DJ, Grace AA: Acute and chronic corticotropin-releasing factor 1 receptor blockade inhibits cocaine-induced dopamine release: correlation with dopamine neuron activity. J. Pharmacol. Exp. Ther.314,201–206 (2005).
  • 154  Kash TL, Nobis WP, Matthews RT, Winder DG: Dopamine enhances fast excitatory synaptic transmission in the extended amygdala by a CRF-R1-dependent process. J. Neurosci.28,13856–13865 (2008).
  • 155  Meloni EG, Gerety LP, Knoll AT, Cohen BM, Carlezon WA Jr: Behavioral and anatomical interactions between dopamine and corticotropin-releasing factor in the rat. J. Neurosci.26,3855–3863 (2006).
  • 156  Rouge-Pont F, Piazza PV, Kharouby M, Le Moal M, Simon H: Higher and longer stress-induced increase in dopamine concentrations in the nucleus accumbens of animals predisposed to amphetamine self-administration. A microdialysis study. Brain Res.602,169–174 (1993).
  • 157  Epstein DH, Preston KL, Stewart J, Shaham Y: Toward a model of drug relapse: an assessment of the validity of the reinstatement procedure. Psychopharmacology (Berl.)189,1–16 (2006).
  • 158  McFarland K, Davidge SB, Lapish CC, Kalivas PW: Limbic and motor circuitry underlying footshock-induced reinstatement of cocaine-seeking behavior. J. Neurosci.24,1551–1560 (2004).
  • 159  Wang B, Shaham Y, Zitzman D et al.: Cocaine experience establishes control of midbrain glutamate and dopamine by corticotropin-releasing factor: a role in stress-induced relapse to drug seeking. J. Neurosci.25,5389–5396 (2005).
  • 160  Mantsch JR, Baker DA, Francis DM et al.: Stressor- and corticotropin releasing factor-induced reinstatement and active stress-related behavioral responses are augmented following long-access cocaine self-administration by rats. Psychopharmacology (Berl.)195,591–603 (2008).
  • 161  Yap JJ, Miczek KA: Stress and rodent models of drug addiction: role of VTA–accumbens–PFC–amygdala circuit. Drug Discov. Today Dis. Models5,259–270 (2008).
  • 162  O’Brien CP: A range of research-based pharmacotherapies for addiction. Science278,66–70 (1997).
  • 163  Ehrman RN, Robbins SJ, Childress AR, O’Brien CP: Conditioned responses to cocaine-related stimuli in cocaine abuse patients. Psychopharmacology (Berl.)107,523–529 (1992).
  • 164  Saladin ME, Brady KT, Graap K, Rothbaum BO: A preliminary report on the use of virtual reality technology to elicit craving and cue reactivity in cocaine dependent individuals. Addict. Behav.31,1881–1894 (2006).
  • 165  Stewart J, de Wit H, Eikelboom R: Role of unconditioned and conditioned drug effects in the self-administration of opiates and stimulants. Psychol. Rev.91,251–268 (1984).
  • 166  Tran-Nguyen LT, Fuchs RA, Coffey GP, Baker DA, O’Dell LE, Neisewander JL: Time-dependent changes in cocaine-seeking behavior and extracellular dopamine levels in the amygdala during cocaine withdrawal. Neuropsychopharmacology19,48–59 (1998).
  • 167  Grimm JW, Hope BT, Wise RA, Shaham Y: Neuroadaptation. Incubation of cocaine craving after withdrawal. Nature412,141–142 (2001).
  • 168  Sinclair JD, Senter RJ: Development of an alcohol-deprivation effect in rats. Q. J. Stud. Alcohol29,863–867 (1968).
  • 169  Spanagel R, Holter SM, Allingham K, Landgraf R, Zieglgansberger W: Acamprosate and alcohol: I. Effects on alcohol intake following alcohol deprivation in the rat. Eur. J. Pharmacol.305,39–44 (1996).
  • 170  Lu L, Grimm JW, Hope BT, Shaham Y: Incubation of cocaine craving after withdrawal: a review of preclinical data. Neuropharmacology47(Suppl. 1),214–226 (2004).
  • 171  Lu L, Hope BT, Dempsey J, Liu SY, Bossert JM, Shaham Y: Central amygdala ERK signaling pathway is critical to incubation of cocaine craving. Nat. Neurosci.8,212–219 (2005).▪▪ Impressive study demonstrating the role of extracellular signal-regulated kinase signaling in the central nucleus of the amygdala in mediating the withdrawal time-dependent increase in cocaine-seeking behavior in rats.
  • 172  Grimm JW, Lu L, Hayashi T, Hope BT, Su TP, Shaham Y: Time-dependent increases in brain-derived neurotrophic factor protein levels within the mesolimbic dopamine system after withdrawal from cocaine: implications for incubation of cocaine craving. J. Neurosci.23,742–747 (2003).
  • 173  Zorrilla EP, Valdez GR, Weiss F: Changes in levels of regional CRF-like-immunoreactivity and plasma corticosterone during protracted drug withdrawal in dependent rats. Psychopharmacology (Berl.)158,374–381 (2001).
  • 174  Siegmund KD, Connor CM, Campan M et al.: DNA methylation in the human cerebral cortex is dynamically regulated throughout the life span and involves differentiated neurons. PLoS ONE2(9),E895 (2007).
  • 175  Chahrour M, Zoghbi HY: The story of Rett syndrome: from clinic to neurobiology. Neuron56,422–437 (2007).
  • 176  Tsankova N, Renthal W, Kumar A, Nestler EJ: Epigenetic regulation in psychiatric disorders. Nat. Rev. Neurosci.8,355–367 (2007).
  • 177  Kumar A, Choi KH, Renthal W et al.: Chromatin remodeling is a key mechanism underlying cocaine-induced plasticity in striatum. Neuron48,303–314 (2005).
  • 178  Graham DL, Edwards S, Bachtell RK, DiLeone RJ, Rios M, Self DW: Dynamic BDNF activity in nucleus accumbens with cocaine use increases self-administration and relapse. Nat. Neurosci.10,1029–1037 (2007).▪ Demonstrates how early drug-induced neuroadaptations can have long-lasting effects on drug-seeking behavior.
  • 179  Renthal W, Maze I, Krishnan V et al.: Histone deacetylase 5 epigenetically controls behavioral adaptations to chronic emotional stimuli. Neuron56,517–529 (2007).
  • 180  Renthal W, Terzi D, Olson EN, Nestler EJ, Zachariou V: Histone deacetylase 5 controls analgesic responsiveness to tricyclic antidepressants in a mouse model of neuropathic pain. Soc. Neurosci. Abstr.34 (2008) (Abstract 368.6).
  • 181  Pandey SC, Ugale R, Zhang H, Tang L, Prakash A: Brain chromatin remodeling: a novel mechanism of alcoholism. J. Neurosci.28,3729–3737 (2008).