Pharmacopsychiatry 2003; 36: 255-258
DOI: 10.1055/s-2003-45139
Original Paper
© Georg Thieme Verlag Stuttgart · New York

Neurobiological Correlates of the Disposition and Maintenance of Alcoholism

A. Heinz1 , M. Schäfer1 , J. D. Higley2 , J. H. Krystal3 , D. Goldman2
  • 1Department of Psychiatry and Psychotherapy, Charité - University Medicine Berlin, Campus Charité-Mitte (CCM), Berlin, Germany
  • 2National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD, USA
  • 3NIAAA Center for the Translational Neuroscience of Alcoholism, Yale University, New Haven, CT, USA
Further Information

Publication History

Publication Date:
15 December 2003 (online)

The last decade witnessed a rapid increase in the knowledge of the etiopathology and treatment of alcoholism. The current disease concept includes psychosocial and neurobiological foundations and consequences of alcoholism. Neurobiological research points to dispositional factors such as a low level of response to alcohol, which is partly heritable and seems to be associated with monoaminergic dysfunction and reduced GABAergic alcohol effects. Chronic alcohol intake stimulates counteradaptive neuroadaptation in central GABAergic and glutamatergic neurotransmission, which increases alcohol tolerance. Neuroadaptation to chronic alcohol effects is not immediately reversed during detoxification and can cause clinical withdrawal once alcohol intake is terminated. Sensitization of the dopaminergic and opioidergic reward system may contribute to alcohol craving and reduced control of alcohol intake. New treatment options include pharmacological approaches and indicate that behavior or motivational therapy and the attendance of patient groups may equally reduce the relapse risk.

References

  • 1 Abi-Dargham A, Krystal J H, Anjilvel S, Scanley B E, Zoghbi S, Baldwin R M,. et al . Alterations of benzodiazepine receptors in type II alcoholic subjects measured with SPECT and [123I]iomazenil.  Am J Psychiatry. 1998;  155 1550-1555
  • 2 Anthony J C, Warner L A, Kessler R C. Comparative epidemiology of dependence on tobacco, alcohol, controlled substances, and inhalants: basic findings from the National Comorbidity Survey.  Exp Clin Psychopharmacology. 1994;  2 244-68
  • 3 Barr C S, Newman T K, Becker M L, Champoux M, Lesch K P, Suomi S J,. et al . Serotonin transporter gene variation is associated with alcohol sensitivity in rhesus macaques exposed to early-life stress.  Alc Clin Exp Res. 2003;  27 812-817
  • 4 Berridge K C, Robinson T E. What is the role of dopamine in reward: hedonic impact, reward learning, or incentive salience?.  Brain Res Rev. 1998;  28 309-369
  • 5 Bien T H, Miller W R, Tonigan S J. Brief interventions for alcohol problems: a review.  Addiction. 1993;  88 315-336
  • 6 Clarke A S, Hedeker D R, Ebert M H, Schmidt D E, McKinney W T, Kraemer G W. Rearing experiments and biogenic amine activity in infant rhesus monkeys.  Biol Psychiat. 1996;  40 338-352
  • 7 Cloninger C R. Neurogenetic adaptive mechanisms in alcoholism.  Science. 1987;  236 410-416
  • 8 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. 1988;  5 5274-5278
  • 9 Di Chiara G. The role of dopamine in drug abuse viewed from the perspective of its role in motivation.  Drug Alcohol Dependence. 1995;  38 95-137
  • 10 Doudet D, Hommer D, Higley J D, Andreason P J, Moneman R, Suomi S S. et al . Cerebral glucose metabolism, CSF 5-HIAA levels, and aggressive behavior in rhesus monkeys.  Am J Psychiat. 1995;  152 1782-1787
  • 11 Engberg G, Hajos M. Alcohol withdrawal reaction as a result of adaptive changes of excitatory amino acid receptors.  Naunyn-Schmiedebach’s Arch Pharmacol. 1992;  346 437-441
  • 12 Heinz A, Higley J D, Jones D W, Gorey J G, Hommer D, Saunders R. et al . In vivo association between brainstem serotonin transporters and sensitivity to alcohol intoxication.  Am J Psychiat. 1998;  155 1023-1028
  • 13 Heinz A, Jones D W, Mazzanti C, Goldman D, Ragan P, Hommer D. et al . A relationship between serotonin transporter genotype and in vivo protein expression and alcohol neurotoxicity.  Biol Psychiatry. 2000;  47 643-649
  • 14 Heinz A, Jones DW; Gorey J, Bennet A, Suomi S J, Linnoila M. et al . Serotonin transporter availability correlates with alcohol intake in non-human primates.  Mol Psychiatry. 2003a;  8 231-234
  • 15 Heinz A, Löber S, Georgi A, Wrase J, Hermann D, Rey E R. et al . Reward craving and withdrawal relief craving: assessment of different motivational pathways to alcohol intake.  Alc Alcohol. 2003b;  38 35-39
  • 16 Higley J D, Suomi S S, Linnoila M. CSF monoamine metabolite concentrations vary according to age, rearing, and sex, and are influenced by the stressor of social separation in rhesus monkeys.  Psychopharmacol (Berl). 1991;  103 551-556
  • 17 Higley J D, Suomi S S, Linnoila M. A non-human primate model of type II excessive alcohol consumption. Part 1 & 2.  Alc Clin Exp Res. 1996;  20 629-651
  • 18 Koob G F, Le Moal M. Drug abuse: hedonic homeostatic dysregulation.  Science. 1997;  278 52-58
  • 19 Krystal J H, Petrakis I L, Webb E, Cooney N L, Karper L P, Namanworth P, Stetson P, Trevisan L A, Charney D S. Dose-related ethanol-like effects of the NMDA antagonist, ketamine, in recently detoxified alcoholics.  Arch Gen Psychiatry. 1998;  55 354-360
  • 20 Krystal J H, Cramer J A, Krol W F, Kirk G F, Rosenheck R A. Naltrexone in the treatment of alcohol dependence.  N Engl J Med. 2001;  345 1734-1739
  • 21 Krystal J H, Tabakoff B. Ethanol abuse, dependence, and withdrawal: neurobiology and clinical implications. In: Davis KL, Charney DS, Coyle JT, Nemeroff C, editors Psychopharmacology: a fifth generation of progress. Philadelphia; Lippincott Williams and Wilkins 2002: p. 1425-43
  • 22 Krystal J H, Petrakis I L, Mason G, Trevisan L, D’Souza D C. N-methyl-D-aspartate glutamate receptors and alcoholism: reward, dependence, treatment, and vulnerability.  Pharmacol Ther. 2003a;  99 79-94
  • 23 Krystal J H, Petrakis I L, Limoncelli D, Webb E, Gueorgueva R, D’Souza D C. et al .Altered NMDA glutamate receptor antagonist response in recovering ethanol dependent patients. Neuropsychopharmacology 2003b (in press)
  • 24 Krupitsky E M, Burakov A M, Romanova T N, Grinenko N I, Grinenko A Y, Fletcher J. et al . Attenuation of ketamine effects by nimodipine pretreatment in recovering ethanol dependent men: psychopharmacologic implications of the interaction of NMDA and L-type calcium channel antagonists.  Neuropsychopharmacology. 2001;  25 936-947
  • 25 Lesch K P, Bengel D, Heils A, Sabol S Z, Greenberg B D, Petri S, Benjamin J, Muller C R, Hamer D H, Murphy D L. Association of anxiety-related traits with a polymorphism in the serotonin transporter gene regulatory region.  Science. 1996;  274 1527-1531
  • 26 Mann K, Hermann D, Heinz A. One hundred years of alcoholism: The twentieth century.  Alc Alcohol. 2000;  35 10-15
  • 27 Mann K, Agartz I, Harper C, Shoaf S, Rawlings R R, Momenan R. et al . Neuroimaging in Alcoholism: Ethanol and Brain Damage.  Alcohol Clin Exp Res. 2001;  25 104S-109S
  • 28 McBride W J, Li T K. Animal models of alcoholism: neurobiology of high alcohol-drinking behavior in rodents.  Crit Rev Neurobiol. 1998;  12 339-369
  • 29 Mihic S J, Ye Q, Wick M J, Koltchine V V, Krasowski M D, Finn S E. et al . Sites of alcohol and volatile anaesthetic action on GABA(A) and glycine receptors.  Nature. 1997;  389 385-389
  • 30 Miller W R, Rollnick S. Motivational interviewing: preparing people to change addictive behavior. Guilford Press New York; 1991
  • 31 Miyakawa T, Yagi T, Kitazawa H, Yasuda M, Kawai N, Tsuboi K. et al . Fyn-kinase as determinant of ethanol sensitivity: relation to NMDA-receptor function.  Science. 1997;  278 698-701
  • 32 Nestler E J. Molecular neurobiology of drug addiction.  Neuropsychopharmacol. 1994;  11 77-87
  • 33 Newlin D B, Thompson J B. Alcohol challenge with sons of alcoholics: a critical review and analysis.  Psychol Bull. 1990;  108 383-402
  • 34 Niaura R S, Rohsenow D J, Binkoff J A, Monti P M, Pedrazza M, Abrams D B. Relevance of cue reactivity to understanding alcohol and smoking relapse.  J Abnormal Psychol. 1988;  97 133-152
  • 35 O’Brien C, Childress A R, Ehrman R, Robbins S J. Conditioning factors in drug abuse: can they explain compulsion?.  J Psychopharmacol. 1998;  12 15-22
  • 36 O’Malley S S, Jaffe A J, Chang G, Rode S, Schottenfeld R, Meyer R E. et al . Six-month follow-up of naltrexone and psychotherapy for alcohol dependence.  Arch Gen Psychiatry. 1996;  53 217-224
  • 37 Oxenstierna G, Edman G, Iselius L, Oreland L, Ross S B, Sedvall G. Concentrations of monoamine metabolism in the cerebrospinal fluid of twins and unrelated individuals - a genetic study.  J Psychiat Res. 1986;  20 19-29
  • 38 Petrakis I P, Limoncelli D, Gueorguieva R, Boutros N N, Trevisan L, Gelernter J. et al .Altered NMDA glutamate receptor antagonist response in individuals with a family vulnerability to alcoholism. Am J Psychiatry 2003 (in press)
  • 39 Pollock V E. Meta-analysis of subjective sensitivity to alcohol in sons of alcoholics.  Am J Psychiatry. 1992;  149 534-1538
  • 40 Sass H, Soyka M, Mann K, Zieglgänsberger W. Relapse prevention by acamprosate: results from a placebo-controlled study on alcohol dependence.  Arch Gen Psychiatry. 1996;  53 673-680
  • 41 Schuckit M A, Smith T L. An 8-year follow-up of 450 sons of alcoholic and control subjects.  Arch Gen Psychiat. 1996;  53 202-210
  • 42 Schuckit M A, Mazzanti C, Smith T L, Ahmed U, Radel M, Iwata N. et al . Selective genotyping for the role of 5-HT2A, 5-HT2C, and GABAα6 receptors and the serotonin transporter in the level of response to alcohol: a pilot study.  Biol Psychiat. 1999;  45 647-651
  • 43 Schultz W, Dayan P, Montague P R. A neural substrate of prediction and reward.  Science. 1997;  275 1593-1599
  • 44 Siegel S, Hinson R E, Krank M D, McCully J. Heroin ”overdose” death: contribution of drug-associated environmental cues.  Science. 1982;  216 436-437
  • 45 Spanagel R, Herz A, Shippenberg T S. Opposing tonically active endogeneous opioid systems modulate the mesolimbic dopaminergic pathway.  Proceed Nat Acad Science USA. 1992;  89 2046-2050
  • 46 Spanagel R, Zieglgänsberger W. Anti-craving compounds for ethanol: new pharmacological tools to study addictive processes.  Trends Pharmacol Sci. 1997;  18 54-59
  • 47 Tsai G, Gastfriend D R, Coyle J T. The glutamatergic basis of human alcoholism.  American Journal of Psychiatry. 1995;  152 332-340
  • 48 Volpicelli J R, Watson N T, King A C, Sherman C E, O’Brien C P. Effect of naltrexone on alcohol ”high” in alcoholics.  Am J Psychiatry. 1995;  152 613-615
  • 49 Wienberg G. Die vergessene Mehrheit. Zur Realität der Versorgung alkohol- und medikamentenabhängiger Patienten. Psychiatrie Verlag Berlin; 1992
  • 50 Wiesbeck G A, Weijers H G, Lesch O M, Glaser T, Toennis P J, Böning J. Flupentixol decanoate and relapse prevention in alcoholics: results from a placebo-controlled study.  Alc Alcoholism. 2001;  36 329-334
  • 51 Wise R A. The neurobiology of craving: implications for the understanding and treatment of addiction.  J Abnormal Psychol. 1988;  97 118-132

Andreas Heinz; MD

Professor of Psychiatry

Director and Chair

Department of Psychiatry and Psychotherapy

Charité - University Medicine Berlin

Campus Charité-Mitte (CCM)

Schumannstr. 20/21

10117 Berlin

Germany

Phone: +49-30-450-517001

Fax: +49-30-450-517921

Email: andreas.heinz@charite.de

URL: http://www.charite.de/psychiatrie

    >