Skip to main content

Advertisement

Log in

Utility of genetically modified mice for understanding the neurobiology of substance use disorders

  • Review Paper
  • Published:
Human Genetics Aims and scope Submit manuscript

Abstract

Advances in our ability to modify the mouse genome have enhanced our understanding of the genetic and neurobiological mechanisms contributing to addiction-related behaviors underlying substance use and abuse. These experimentally induced manipulations permit greater spatial and temporal specificity for modification of gene expression within specific cellular populations and during select developmental time periods. In this review, we consider the current mouse genetic model systems that have been employed to understand aspects of addiction and highlight significant conceptual advances achieved related to substance use and abuse. The mouse models reviewed herein include conventional knockout and knockin, conditional knockout, transgenic, inducible transgenic, mice suitable for optogenetic control of discrete neuronal populations, and phenotype-selected mice. By establishing a reciprocal investigatory relationship between genetic findings in humans and genomic manipulations in mice, a far better understanding of the discrete neuromechanisms underlying addiction can be achieved, which is likely to provide a strong foundation for developing and validating novel therapeutics for the treatment of substance abuse disorders.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  • Adamantidis AR, Tsai HC, Boutrel B, Zhang F, Stuber GD, Budygin EA, Tourino C, Bonci A, Deisseroth K, de Lecea L (2011) Optogenetic interrogation of dopaminergic modulation of the multiple phases of reward-seeking behavior. J Neurosci 31:10829–10835

    PubMed  CAS  Google Scholar 

  • Agatsuma S, Dang MT, Li Y, Hiroi N (2010) N-methyl-d-aspartic acid receptors on striatal neurons are essential for cocaine cue reactivity in mice. Biol Psychiatry 67:778–780

    PubMed  CAS  Google Scholar 

  • Akbarian S, Bates B, Liu RJ, Skirboll SL, Pejchal T, Coppola V, Sun LD, Fan G, Kucera J, Wilson MA, Tessarollo L, Kosofsky BE, Taylor JR, Bothwell M, Nestler EJ, Aghajanian GK, Jaenisch R (2001) Neurotrophin-3 modulates noradrenergic neuron function and opiate withdrawal. Mol Psychiatry 6:593–604

    PubMed  CAS  Google Scholar 

  • Akbarian S, Rios M, Liu RJ, Gold SJ, Fong HF, Zeiler S, Coppola V, Tessarollo L, Jones KR, Nestler EJ, Aghajanian GK, Jaenisch R (2002) Brain-derived neurotrophic factor is essential for opiate-induced plasticity of noradrenergic neurons. J Neurosci 22:4153–4162

    PubMed  CAS  Google Scholar 

  • Aylor DL, Valdar W, Foulds-Mathes W, Buus RJ, Verdugo RA, Baric RS, Ferris MT, Frelinger JA, Heise M, Frieman MB, Gralinski LE, Bell TA, Didion JD, Hua K, Nehrenberg DL, Powell CL, Steigerwalt J, Xie Y, Kelada SN, Collins FS, Yang IV, Schwartz DA, Branstetter LA, Chesler EJ, Miller DR, Spence J, Liu EY, McMillan L, Sarkar A, Wang J, Wang W, Zhang Q, Broman KW, Korstanje R, Durrant C, Mott R, Iraqi FA, Pomp D, Threadgill D, Pardo-Manuel de Villena F, Churchill GA (2011) Genetic analysis of complex traits in the emerging collaborative cross. Genome Res 21:1213–1222

    Google Scholar 

  • Azizian A, Monterosso J, O’Neill J, London ED (2009) Magnetic resonance imaging studies of cigarette smoking. Handb Exp Pharmacol 192:113–143

    Google Scholar 

  • Azizian A, Nestor LJ, Payer D, Monterosso JR, Brody AL, London ED (2010) Smoking reduces conflict-related anterior cingulate activity in abstinent cigarette smokers performing a Stroop task. Neuropsychopharmacology 35:775–782

    PubMed  Google Scholar 

  • Bastia E, Xu YH, Scibelli AC, Day YJ, Linden J, Chen JF, Schwarzschild MA (2005) A crucial role for forebrain adenosine A(2A) receptors in amphetamine sensitization. Neuropsychopharmacology 30:891–900

    PubMed  CAS  Google Scholar 

  • Batel P, Houchi H, Daoust M, Ramoz N, Naassila M, Gorwood P (2008) A haplotype of the DRD1 gene is associated with alcohol dependence. Alcohol Clin Exp Res 32:567–572

    PubMed  CAS  Google Scholar 

  • Bello EP, Mateo Y, Gelman DM, Noain D, Shin JH, Low MJ, Alvarez VA, Lovinger DM, Rubinstein M (2011) Cocaine supersensitivity and enhanced motivation for reward in mice lacking dopamine D(2) autoreceptors. Nat Neurosci 14:1033–1038

    PubMed  CAS  Google Scholar 

  • Berrettini W, Yuan X, Tozzi F, Song K, Francks C, Chilcoat H, Waterworth D, Muglia P, Mooser V (2008) Alpha-5/alpha-3 nicotinic receptor subunit alleles increase risk for heavy smoking. Mol Psychiatry 13:368–373

    PubMed  CAS  Google Scholar 

  • Berton O, McClung CA, Dileone RJ, Krishnan V, Renthal W, Russo SJ, Graham D, Tsankova NM, Bolanos CA, Rios M, Monteggia LM, Self DW, Nestler EJ (2006) Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science 311:864–868

    PubMed  CAS  Google Scholar 

  • Beutler LR, Wanat MJ, Quintana A, Sanz E, Bamford NS, Zweifel LS, Palmiter RD (2011) Balanced NMDA receptor activity in dopamine D1 receptor (D1R)- and D2R-expressing medium spiny neurons is required for amphetamine sensitization. Proc Natl Acad Sci USA 108:4206–4211

    PubMed  CAS  Google Scholar 

  • Bierut LJ, Stitzel JA, Wang JC, Hinrichs AL, Grucza RA, Xuei X, Saccone NL, Saccone SF, Bertelsen S, Fox L, Horton WJ, Breslau N, Budde J, Cloninger CR, Dick DM, Foroud T, Hatsukami D, Hesselbrock V, Johnson EO, Kramer J, Kuperman S, Madden PA, Mayo K, Nurnberger J Jr, Pomerleau O, Porjesz B, Reyes O, Schuckit M, Swan G, Tischfield JA, Edenberg HJ, Rice JP, Goate AM (2008) Variants in nicotinic receptors and risk for nicotine dependence. Am J Psychiatry 165:1163–1171

    PubMed  Google Scholar 

  • Bilbao A, Parkitna JR, Engblom D, Perreau-Lenz S, Sanchis-Segura C, Schneider M, Konopka W, Westphal M, Breen G, Desrivieres S, Klugmann M, Guindalini C, Vallada H, Laranjeira R, de Fonseca FR, Schumann G, Schutz G, Spanagel R (2008) Loss of the Ca2+/calmodulin-dependent protein kinase type IV in dopaminoceptive neurons enhances behavioral effects of cocaine. Proc Natl Acad Sci USA 105:17549–17554

    PubMed  CAS  Google Scholar 

  • Blomqvist O, Gelernter J, Kranzler HR (2000) Family-based study of DRD2 alleles in alcohol and drug dependence. Am J Med Genet 96:659–664

    PubMed  CAS  Google Scholar 

  • Boer U, Noll C, Cierny I, Krause D, Hiemke C, Knepel W (2010) A common mechanism of action of the selective serotonin reuptake inhibitors citalopram and fluoxetine: reversal of chronic psychosocial stress-induced increase in CRE/CREB-directed gene transcription in transgenic reporter gene mice. Eur J Pharmacol 633:33–38

    PubMed  Google Scholar 

  • Bolla K, Ernst M, Kiehl K, Mouratidis M, Eldreth D, Contoreggi C, Matochik J, Kurian V, Cadet J, Kimes A, Funderburk F, London E (2004) Prefrontal cortical dysfunction in abstinent cocaine abusers. J Neuropsychiatry Clin Neurosci 16:456–464

    PubMed  Google Scholar 

  • Brody AL, Mandelkern MA, London ED, Olmstead RE, Farahi J, Scheibal D, Jou J, Allen V, Tiongson E, Chefer SI, Koren AO, Mukhin AG (2006a) Cigarette smoking saturates brain alpha 4 beta 2 nicotinic acetylcholine receptors. Arch Gen Psychiatry 63:907–915

    PubMed  CAS  Google Scholar 

  • Brody AL, Mandelkern MA, Olmstead RE, Scheibal D, Hahn E, Shiraga S, Zamora-Paja E, Farahi J, Saxena S, London ED, McCracken JT (2006b) Gene variants of brain dopamine pathways and smoking-induced dopamine release in the ventral caudate/nucleus accumbens. Arch Gen Psychiatry 63:808–816

    PubMed  CAS  Google Scholar 

  • Cahir E, Pillidge K, Drago J, Lawrence AJ (2011) The necessity of alpha4* nicotinic receptors in nicotine-driven behaviors: dissociation between reinforcing and motor effects of nicotine. Neuropsychopharmacology 36:1505–1517

    PubMed  CAS  Google Scholar 

  • Chen R, Tilley MR, Wei H, Zhou F, Zhou FM, Ching S, Quan N, Stephens RL, Hill ER, Nottoli T, Han DD, Gu HH (2006) Abolished cocaine reward in mice with a cocaine-insensitive dopamine transporter. Proc Natl Acad Sci USA 103:9333–9338

    PubMed  CAS  Google Scholar 

  • Chen AA, Thomas DK, Ong LL, Schwartz RE, Golub TR, Bhatia SN (2011) Humanized mice with ectopic artificial liver tissues. Proc Natl Acad Sci USA 108:11842–11847

    Google Scholar 

  • Chuhma N, Tanaka KF, Hen R, Rayport S (2011) Functional connectome of the striatal medium spiny neuron. J Neurosci 31:1183–1192

    PubMed  CAS  Google Scholar 

  • Colby CR, Whisler K, Steffen C, Nestler EJ, Self DW (2003) Striatal cell type-specific overexpression of DeltaFosB enhances incentive for cocaine. J Neurosci 23:2488–2493

    PubMed  CAS  Google Scholar 

  • Collins FS, Rossant J, Wurst W (2007) A mouse for all reasons. Cell 128:9–13

    PubMed  CAS  Google Scholar 

  • Corbille AG, Valjent E, Marsicano G, Ledent C, Lutz B, Herve D, Girault JA (2007) Role of cannabinoid type 1 receptors in locomotor activity and striatal signaling in response to psychostimulants. J Neurosci 27:6937–6947

    PubMed  CAS  Google Scholar 

  • Crabbe JC, Phillips TJ, Harris RA, Arends MA, Koob GF (2006) Alcohol-related genes: contributions from studies with genetically engineered mice. Addict Biol 11:195–269

    PubMed  CAS  Google Scholar 

  • Dahl JP, Weller AE, Kampman KM, Oslin DW, Lohoff FW, Ferraro TN, O’Brien CP, Berrettini WH (2005) Confirmation of the association between a polymorphism in the promoter region of the prodynorphin gene and cocaine dependence. Am J Med Genet B Neuropsychiatr Genet 139B:106–108

    PubMed  Google Scholar 

  • Darvas M, Fadok JP, Palmiter RD (2011) Requirement of dopamine signaling in the amygdala and striatum for learning and maintenance of a conditioned avoidance response. Learn Mem 18:136–143

    PubMed  CAS  Google Scholar 

  • De Biasi M, Dani JA (2011) Reward, addiction, withdrawal to nicotine. Annu Rev Neurosci 34:105–130

    PubMed  Google Scholar 

  • Deroche-Gamonet V, Sillaber I, Aouizerate B, Izawa R, Jaber M, Ghozland S, Kellendonk C, Le Moal M, Spanagel R, Schutz G, Tronche F, Piazza PV (2003) The glucocorticoid receptor as a potential target to reduce cocaine abuse. J Neurosci 23:4785–4790

    PubMed  CAS  Google Scholar 

  • Dinieri JA, Nemeth CL, Parsegian A, Carle T, Gurevich VV, Gurevich E, Neve RL, Nestler EJ, Carlezon WA Jr (2009) Altered sensitivity to rewarding and aversive drugs in mice with inducible disruption of cAMP response element-binding protein function within the nucleus accumbens. J Neurosci 29:1855–1859

    PubMed  CAS  Google Scholar 

  • Domenici MR, Azad SC, Marsicano G, Schierloh A, Wotjak CT, Dodt HU, Zieglgansberger W, Lutz B, Rammes G (2006) Cannabinoid receptor type 1 located on presynaptic terminals of principal neurons in the forebrain controls glutamatergic synaptic transmission. J Neurosci 26:5794–5799

    PubMed  CAS  Google Scholar 

  • Donovan DM, Miner LL, Perry MP, Revay RS, Sharpe LG, Przedborski S, Kostic V, Philpot RM, Kirstein CL, Rothman RB, Schindler CW, Uhl GR (1999) Cocaine reward and MPTP toxicity: alteration by regional variant dopamine transporter overexpression. Brain Res Mol Brain Res 73:37–49

    PubMed  CAS  Google Scholar 

  • Doyle JP, Dougherty JD, Heiman M, Schmidt EF, Stevens TR, Ma G, Bupp S, Shrestha P, Shah RD, Doughty ML, Gong S, Greengard P, Heintz N (2008) Application of a translational profiling approach for the comparative analysis of CNS cell types. Cell 135:749–762

    PubMed  CAS  Google Scholar 

  • Drenan RM, Grady SR, Whiteaker P, McClure-Begley T, McKinney S, Miwa JM, Bupp S, Heintz N, McIntosh JM, Bencherif M, Marks MJ, Lester HA (2008) In vivo activation of midbrain dopamine neurons via sensitized, high-affinity alpha 6 nicotinic acetylcholine receptors. Neuron 60:123–136

    PubMed  CAS  Google Scholar 

  • Drenan RM, Grady SR, Steele AD, McKinney S, Patzlaff NE, McIntosh JM, Marks MJ, Miwa JM, Lester HA (2010) Cholinergic modulation of locomotion and striatal dopamine release is mediated by alpha6alpha4* nicotinic acetylcholine receptors. J Neurosci 30:9877–9889

    PubMed  CAS  Google Scholar 

  • Dymecki SM, Ray RS, Kim JC (2010) Mapping cell fate and function using recombinase-based intersectional strategies. Methods Enzymol 477:183–213

    PubMed  CAS  Google Scholar 

  • Engblom D, Bilbao A, Sanchis-Segura C, Dahan L, Perreau-Lenz S, Balland B, Parkitna JR, Lujan R, Halbout B, Mameli M, Parlato R, Sprengel R, Luscher C, Schutz G, Spanagel R (2008) Glutamate receptors on dopamine neurons control the persistence of cocaine seeking. Neuron 59:497–508

    PubMed  CAS  Google Scholar 

  • Ersche KD, Fletcher PC, Lewis SJ, Clark L, Stocks-Gee G, London M, Deakin JB, Robbins TW, Sahakian BJ (2005) Abnormal frontal activations related to decision-making in current and former amphetamine and opiate dependent individuals. Psychopharmacology (Berl) 180:612–623

    CAS  Google Scholar 

  • Exley R, Maubourguet N, David V, Eddine R, Evrard A, Pons S, Marti F, Threlfell S, Cazala P, McIntosh JM, Changeux JP, Maskos U, Cragg SJ, Faure P (2011) Distinct contributions of nicotinic acetylcholine receptor subunit alpha4 and subunit alpha6 to the reinforcing effects of nicotine. Proc Natl Acad Sci USA 108:7577–7582

    PubMed  CAS  Google Scholar 

  • Fowler CD, Kenny PJ (2011) Intravenous nicotine self-administration and cue-induced reinstatement in mice: effects of nicotine dose, rate of drug infusion and prior instrumental training. Neuropharmacology 61:687–698

    PubMed  CAS  Google Scholar 

  • Fowler CD, Lu Q, Johnson PM, Marks MJ, Kenny PJ (2011) Habenular alpha5 nicotinic receptor subunit signalling controls nicotine intake. Nature 471:597–601

    PubMed  CAS  Google Scholar 

  • Frahm S, Slimak MA, Ferrarese L, Santos-Torres J, Antolin-Fontes B, Auer S, Filkin S, Pons S, Fontaine JF, Tsetlin V, Maskos U, Ibanez-Tallon I (2011) Aversion to nicotine is regulated by the balanced activity of beta4 and alpha5 nicotinic receptor subunits in the medial habenula. Neuron 70:522–535

    PubMed  CAS  Google Scholar 

  • Gelernter J, Panhuysen C, Weiss R, Brady K, Hesselbrock V, Rounsaville B, Poling J, Wilcox M, Farrer L, Kranzler HR (2005) Genomewide linkage scan for cocaine dependence and related traits: significant linkages for a cocaine-related trait and cocaine-induced paranoia. Am J Med Genet B Neuropsychiatr Genet 136B:45–52

    PubMed  Google Scholar 

  • Gelernter J, Panhuysen C, Wilcox M, Hesselbrock V, Rounsaville B, Poling J, Weiss R, Sonne S, Zhao H, Farrer L, Kranzler HR (2006) Genomewide linkage scan for opioid dependence and related traits. Am J Hum Genet 78:759–769

    PubMed  CAS  Google Scholar 

  • GENSAT (2011) The Gene Expression Nervous System Atlas (GENSAT) Project, NINDS Contracts N01NS02331 & HHSN271200723701C. The Rockefeller University, New York

  • Gertler TS, Chan CS, Surmeier DJ (2008) Dichotomous anatomical properties of adult striatal medium spiny neurons. J Neurosci 28:10814–10824

    PubMed  CAS  Google Scholar 

  • Giraldo P, Montoliu L (2001) Size matters: use of YACs, BACs and PACs in transgenic animals. Transgenic Res 10:83–103

    PubMed  CAS  Google Scholar 

  • Gong S, Doughty M, Harbaugh CR, Cummins A, Hatten ME, Heintz N, Gerfen CR (2007) Targeting Cre recombinase to specific neuron populations with bacterial artificial chromosome constructs. J Neurosci 27:9817–9823

    PubMed  CAS  Google Scholar 

  • Graham DL, Krishnan V, Larson EB, Graham A, Edwards S, Bachtell RK, Simmons D, Gent LM, Berton O, Bolanos CA, DiLeone RJ, Parada LF, Nestler EJ, Self DW (2009) Tropomyosin-related kinase B in the mesolimbic dopamine system: region-specific effects on cocaine reward. Biol Psychiatry 65:696–701

    PubMed  CAS  Google Scholar 

  • Guan C, Ye C, Yang X, Gao J (2010) A review of current large-scale mouse knockout efforts. Genesis 48:73–85

    PubMed  CAS  Google Scholar 

  • Hashimoto JG, Forquer MR, Tanchuck MA, Finn DA, Wiren KM (2011) Importance of genetic background for risk of relapse shown in altered prefrontal cortex gene expression during abstinence following chronic alcohol intoxication. Neuroscience 173:57–75

    PubMed  CAS  Google Scholar 

  • Heiman M, Schaefer A, Gong S, Peterson JD, Day M, Ramsey KE, Suarez-Farinas M, Schwarz C, Stephan DA, Surmeier DJ, Greengard P, Heintz N (2008) A translational profiling approach for the molecular characterization of CNS cell types. Cell 135:738–748

    PubMed  CAS  Google Scholar 

  • Heintz N (2001) BAC to the future: the use of bac transgenic mice for neuroscience research. Nat Rev Neurosci 2:861–870

    PubMed  CAS  Google Scholar 

  • Hendrickson LM, Zhao-Shea R, Pang X, Gardner PD, Tapper AR (2010) Activation of alpha4* nAChRs is necessary and sufficient for varenicline-induced reduction of alcohol consumption. J Neurosci 30:10169–10176

    PubMed  CAS  Google Scholar 

  • Heyer MP, Feliciano C, Peça J, Feng G (2011) Elucidating gene function through use of genetically engineered mice. In: Starkey MPE, Elaswarapu R (eds) Genomics: essential methods. Wiley-Blackwell, Oxford, pp 211–248

    Google Scholar 

  • Hikida T, Kimura K, Wada N, Funabiki K, Nakanishi S (2010) Distinct roles of synaptic transmission in direct and indirect striatal pathways to reward and aversive behavior. Neuron 66:896–907

    PubMed  CAS  Google Scholar 

  • Hitz C, Steuber-Buchberger P, Delic S, Wurst W, Kuhn R (2009) Generation of shRNA transgenic mice. Methods Mol Biol 530:101–129

    PubMed  CAS  Google Scholar 

  • Hnasko TS, Perez FA, Scouras AD, Stoll EA, Gale SD, Luquet S, Phillips PE, Kremer EJ, Palmiter RD (2006) Cre recombinase-mediated restoration of nigrostriatal dopamine in dopamine-deficient mice reverses hypophagia and bradykinesia. Proc Natl Acad Sci USA 103:8858–8863

    PubMed  CAS  Google Scholar 

  • Hnasko TS, Sotak BN, Palmiter RD (2007) Cocaine-conditioned place preference by dopamine-deficient mice is mediated by serotonin. J Neurosci 27:12484–12488

    PubMed  CAS  Google Scholar 

  • Hnasko TS, Chuhma N, Zhang H, Goh GY, Sulzer D, Palmiter RD, Rayport S, Edwards RH (2010) Vesicular glutamate transport promotes dopamine storage and glutamate corelease in vivo. Neuron 65:643–656

    PubMed  CAS  Google Scholar 

  • Hollander JA, Im HI, Amelio AL, Kocerha J, Bali P, Lu Q, Willoughby D, Wahlestedt C, Conkright MD, Kenny PJ (2010) Striatal microRNA controls cocaine intake through CREB signalling. Nature 466:197–202

    PubMed  CAS  Google Scholar 

  • Hummel M, Ansonoff MA, Pintar JE, Unterwald EM (2004) Genetic and pharmacological manipulation of mu opioid receptors in mice reveals a differential effect on behavioral sensitization to cocaine. Neuroscience 125:211–220

    PubMed  CAS  Google Scholar 

  • Im HI, Hollander JA, Bali P, Kenny PJ (2010) MeCP2 controls BDNF expression and cocaine intake through homeostatic interactions with microRNA-212. Nat Neurosci 13:1120–1127

    PubMed  CAS  Google Scholar 

  • International Mouse Knockout Consortium (2007) A mouse for all reasons. Cell 128:9–13

    Google Scholar 

  • Irion S, Luche H, Gadue P, Fehling HJ, Kennedy M, Keller G (2007) Identification and targeting of the ROSA26 locus in human embryonic stem cells. Nat Biotechnol 25:1477–1482

    PubMed  CAS  Google Scholar 

  • Izawa R, Jaber M, Deroche-Gamonet V, Sillaber I, Kellendonk C, Le Moal M, Tronche F, Piazza PV (2006) Gene expression regulation following behavioral sensitization to cocaine in transgenic mice lacking the glucocorticoid receptor in the brain. Neuroscience 137:915–924

    PubMed  CAS  Google Scholar 

  • Jeon J, Dencker D, Wortwein G, Woldbye DP, Cui Y, Davis AA, Levey AI, Schutz G, Sager TN, Mork A, Li C, Deng CX, Fink-Jensen A, Wess J (2010) A subpopulation of neuronal M4 muscarinic acetylcholine receptors plays a critical role in modulating dopamine-dependent behaviors. J Neurosci 30:2396–2405

    PubMed  CAS  Google Scholar 

  • Katzel D, Zemelman BV, Buetfering C, Wolfel M, Miesenbock G (2011) The columnar and laminar organization of inhibitory connections to neocortical excitatory cells. Nat Neurosci 14:100–107

    PubMed  Google Scholar 

  • Kelz MB, Chen J, Carlezon WA Jr, Whisler K, Gilden L, Beckmann AM, Steffen C, Zhang YJ, Marotti L, Self DW, Tkatch T, Baranauskas G, Surmeier DJ, Neve RL, Duman RS, Picciotto MR, Nestler EJ (1999) Expression of the transcription factor deltaFosB in the brain controls sensitivity to cocaine. Nature 401:272–276

    PubMed  CAS  Google Scholar 

  • King SL, Marks MJ, Grady SR, Caldarone BJ, Koren AO, Mukhin AG, Collins AC, Picciotto MR (2003) Conditional expression in corticothalamic efferents reveals a developmental role for nicotinic acetylcholine receptors in modulation of passive avoidance behavior. J Neurosci 23:3837–3843

    PubMed  CAS  Google Scholar 

  • Koentgen F, Suess G, Naf D (2010) Engineering the mouse genome to model human disease for drug discovery. Methods Mol Biol 602:55–77

    PubMed  CAS  Google Scholar 

  • Konopka W, Kiryk A, Novak M, Herwerth M, Parkitna JR, Wawrzyniak M, Kowarsch A, Michaluk P, Dzwonek J, Arnsperger T, Wilczynski G, Merkenschlager M, Theis FJ, Kohr G, Kaczmarek L, Schutz G (2010) MicroRNA loss enhances learning and memory in mice. J Neurosci 30:14835–14842

    PubMed  CAS  Google Scholar 

  • Kosobud A, Crabbe JC (1986) Ethanol withdrawal in mice bred to be genetically prone or resistant to ethanol withdrawal seizures. J Pharmacol Exp Ther 238:170–177

    PubMed  CAS  Google Scholar 

  • Kramer PF, Christensen CH, Hazelwood LA, Dobi A, Bock R, Sibley DR, Mateo Y, Alvarez VA (2011) Dopamine D2 receptor overexpression alters behavior and physiology in Drd2-EGFP mice. J Neurosci 31:126–132

    PubMed  CAS  Google Scholar 

  • Kranz A, Fu J, Duerschke K, Weidlich S, Naumann R, Stewart AF, Anastassiadis K (2010) An improved Flp deleter mouse in C57Bl/6 based on Flpo recombinase. Genesis 48:512–520

    PubMed  CAS  Google Scholar 

  • Kreek MJ, Bart G, Lilly C, LaForge KS, Nielsen DA (2005) Pharmacogenetics and human molecular genetics of opiate and cocaine addictions and their treatments. Pharmacol Rev 57:1–26

    PubMed  CAS  Google Scholar 

  • Kreitzer AC, Malenka RC (2007) Endocannabinoid-mediated rescue of striatal LTD and motor deficits in Parkinson’s disease models. Nature 445:643–647

    PubMed  CAS  Google Scholar 

  • Lambert R (2009) JAX mice research tool strains. The Jackson Laboratory, Bar Harbor

  • Lee KW, Kim Y, Kim AM, Helmin K, Nairn AC, Greengard P (2006) Cocaine-induced dendritic spine formation in D1 and D2 dopamine receptor-containing medium spiny neurons in nucleus accumbens. Proc Natl Acad Sci USA 103:3399–3404

    PubMed  CAS  Google Scholar 

  • Lewandoski M, Martin GR (1997) Cre-mediated chromosome loss in mice. Nat Genet 17:223–225

    PubMed  CAS  Google Scholar 

  • Lobo MK, Covington HE 3rd, Chaudhury D, Friedman AK, Sun H, Damez-Werno D, Dietz DM, Zaman S, Koo JW, Kennedy PJ, Mouzon E, Mogri M, Neve RL, Deisseroth K, Han MH, Nestler EJ (2010) Cell type-specific loss of BDNF signaling mimics optogenetic control of cocaine reward. Science 330:385–390

    PubMed  CAS  Google Scholar 

  • London ED, Cascella NG, Wong DF, Phillips RL, Dannals RF, Links JM, Herning R, Grayson R, Jaffe JH, Wagner HN Jr (1990) Cocaine-induced reduction of glucose utilization in human brain. A study using positron emission tomography and [fluorine 18]-fluorodeoxyglucose. Arch Gen Psychiatry 47:567–574

    PubMed  CAS  Google Scholar 

  • Lu Y, Wu D, Wang X, Ward SC, Cederbaum AI (2010) Chronic alcohol-induced liver injury and oxidant stress are decreased in cytochrome P4502E1 knockout mice and restored in humanized cytochrome P4502E1 knock-in mice. Free Radic Biol Med 49:1406–1416

    PubMed  CAS  Google Scholar 

  • Luo X, Kranzler HR, Zuo L, Yang BZ, Lappalainen J, Gelernter J (2005) ADH4 gene variation is associated with alcohol and drug dependence: results from family controlled and population-structured association studies. Pharmacogenet Genomics 15:755–768

    PubMed  CAS  Google Scholar 

  • Luo Y, Good CH, Diaz-Ruiz O, Zhang Y, Hoffman AF, Shan L, Kuang SY, Malik N, Chefer VI, Tomac AC, Lupica CR, Backman CM (2010) NMDA receptors on non-dopaminergic neurons in the VTA support cocaine sensitization. PLoS One 5:e12141

    PubMed  Google Scholar 

  • Mague SD, Blendy JA (2010) OPRM1 SNP (A118G): involvement in disease development, treatment response, and animal models. Drug Alcohol Depend 108:172–182

    PubMed  CAS  Google Scholar 

  • Mague SD, Isiegas C, Huang P, Liu-Chen LY, Lerman C, Blendy JA (2009) Mouse model of OPRM1 (A118G) polymorphism has sex-specific effects on drug-mediated behavior. Proc Natl Acad Sci USA 106:10847–10852

    PubMed  CAS  Google Scholar 

  • Marks MJ, Pauly JR, Gross SD, Deneris ES, Hermans-Borgmeyer I, Heinemann SF, Collins AC (1992) Nicotine binding and nicotinic receptor subunit RNA after chronic nicotine treatment. J Neurosci 12:2765–2784

    PubMed  CAS  Google Scholar 

  • Maze I, Covington HE 3rd, Dietz DM, LaPlant Q, Renthal W, Russo SJ, Mechanic M, Mouzon E, Neve RL, Haggarty SJ, Ren Y, Sampath SC, Hurd YL, Greengard P, Tarakhovsky A, Schaefer A, Nestler EJ (2010) Essential role of the histone methyltransferase G9a in cocaine-induced plasticity. Science 327:213–216

    PubMed  CAS  Google Scholar 

  • McGranahan TM, Patzlaff NE, Grady SR, Heinemann SF, Booker TK (2011) {alpha}4{beta}2 nicotinic acetylcholine receptors on dopaminergic neurons mediate nicotine reward and anxiety relief. J Neurosci 31:10891–10902

    PubMed  CAS  Google Scholar 

  • McGraw CM, Samaco RC, Zoghbi HY (2011) Adult neural function requires MeCP2. Science 333:186

    PubMed  CAS  Google Scholar 

  • McPherson CS, Mantamadiotis T, Tan SS, Lawrence AJ (2010) Deletion of CREB1 from the dorsal telencephalon reduces motivational properties of cocaine. Cereb Cortex 20:941–952

    PubMed  Google Scholar 

  • Meijer AH, Ouwerkerk PB, Hoge JH (1998) Vectors for transcription factor cloning and target site identification by means of genetic selection in yeast. Yeast 14:1407–1415

    PubMed  CAS  Google Scholar 

  • Mineur YS, Abizaid A, Rao Y, Salas R, DiLeone RJ, Gundisch D, Diano S, De Biasi M, Horvath TL, Gao XB, Picciotto MR (2011) Nicotine decreases food intake through activation of POMC neurons. Science 332:1330–1332

    PubMed  CAS  Google Scholar 

  • Minichiello L, Korte M, Wolfer D, Kuhn R, Unsicker K, Cestari V, Rossi-Arnaud C, Lipp HP, Bonhoeffer T, Klein R (1999) Essential role for TrkB receptors in hippocampus-mediated learning. Neuron 24:401–414

    PubMed  CAS  Google Scholar 

  • Monory K, Blaudzun H, Massa F, Kaiser N, Lemberger T, Schutz G, Wotjak CT, Lutz B, Marsicano G (2007) Genetic dissection of behavioural and autonomic effects of Delta(9)-tetrahydrocannabinol in mice. PLoS Biol 5:e269

    PubMed  Google Scholar 

  • Morice E, Denis C, Giros B, Nosten-Bertrand M (2004) Phenotypic expression of the targeted null-mutation in the dopamine transporter gene varies as a function of the genetic background. Eur J Neurosci 20:120–126

    PubMed  Google Scholar 

  • Nagy A (2000) Cre recombinase: the universal reagent for genome tailoring. Genesis 26:99–109

    PubMed  CAS  Google Scholar 

  • Newton SS, Thome J, Wallace TL, Shirayama Y, Schlesinger L, Sakai N, Chen J, Neve R, Nestler EJ, Duman RS (2002) Inhibition of cAMP response element-binding protein or dynorphin in the nucleus accumbens produces an antidepressant-like effect. J Neurosci 22:10883–10890

    PubMed  CAS  Google Scholar 

  • Novak M, Halbout B, O’Connor EC, Rodriguez Parkitna J, Su T, Chai M, Crombag HS, Bilbao A, Spanagel R, Stephens DN, Schutz G, Engblom D (2010) Incentive learning underlying cocaine-seeking requires mGluR5 receptors located on dopamine D1 receptor-expressing neurons. J Neurosci 30:11973–11982

    PubMed  CAS  Google Scholar 

  • Odell J, Caimi P, Sauer B, Russell S (1990) Site-directed recombination in the genome of transgenic tobacco. Mol Gen Genet 223:369–378

    PubMed  CAS  Google Scholar 

  • Parker J, Beutler L, Palmiter R (2011) The contribution of NMDA receptor signaling in the corticobasal ganglia reward network to appetitive pavlovian learning. J Neurosci 31:11362–11369

    PubMed  CAS  Google Scholar 

  • Pletnikov MV, Ayhan Y, Nikolskaia O, Xu Y, Ovanesov MV, Huang H, Mori S, Moran TH, Ross CA (2008) Inducible expression of mutant human DISC1 in mice is associated with brain and behavioral abnormalities reminiscent of schizophrenia. Mol Psychiatry 13: 173–186, 115

    Google Scholar 

  • Ray RS, Corcoran AE, Brust RD, Kim JC, Richerson GB, Nattie E, Dymecki SM (2011) Impaired respiratory and body temperature control upon acute serotonergic neuron inhibition. Science 333:637–642

    PubMed  CAS  Google Scholar 

  • Ren J, Qin C, Hu F, Tan J, Qiu L, Zhao S, Feng G, Luo M (2011) Habenula “cholinergic” neurons co-release glutamate and acetylcholine and activate postsynaptic neurons via distinct transmission modes. Neuron 69:445–452

    PubMed  CAS  Google Scholar 

  • Rios M, Lambe EK, Liu R, Teillon S, Liu J, Akbarian S, Roffler-Tarlov S, Jaenisch R, Aghajanian GK (2006) Severe deficits in 5-HT2A -mediated neurotransmission in BDNF conditional mutant mice. J Neurobiol 66:408–420

    PubMed  CAS  Google Scholar 

  • Robinson S, Rainwater AJ, Hnasko TS, Palmiter RD (2007) Viral restoration of dopamine signaling to the dorsal striatum restores instrumental conditioning to dopamine-deficient mice. Psychopharmacology (Berl) 191:567–578

    CAS  Google Scholar 

  • Rudolph D, Tafuri A, Gass P, Hammerling GJ, Arnold B, Schutz G (1998) Impaired fetal T cell development and perinatal lethality in mice lacking the cAMP response element binding protein. Proc Natl Acad Sci USA 95:4481–4486

    PubMed  CAS  Google Scholar 

  • Russo SJ, Wilkinson MB, Mazei-Robison MS, Dietz DM, Maze I, Krishnan V, Renthal W, Graham A, Birnbaum SG, Green TA, Robison B, Lesselyong A, Perrotti LI, Bolanos CA, Kumar A, Clark MS, Neumaier JF, Neve RL, Bhakar AL, Barker PA, Nestler EJ (2009) Nuclear factor kappa B signaling regulates neuronal morphology and cocaine reward. J Neurosci 29:3529–3537

    PubMed  CAS  Google Scholar 

  • Sacca R, Engle SJ, Qin W, Stock JL, McNeish JD (2010) Genetically engineered mouse models in drug discovery research. Methods Mol Biol 602:37–54

    PubMed  CAS  Google Scholar 

  • Sakai N, Thome J, Newton SS, Chen J, Kelz MB, Steffen C, Nestler EJ, Duman RS (2002) Inducible and brain region-specific CREB transgenic mice. Mol Pharmacol 61:1453–1464

    PubMed  CAS  Google Scholar 

  • Sauer B (1998) Inducible gene targeting in mice using the Cre/lox system. Methods 14:381–392

    PubMed  CAS  Google Scholar 

  • Sauer B, Henderson N (1988) Site-specific DNA recombination in mammalian cells by the Cre recombinase of bacteriophage P1. Proc Natl Acad Sci USA 85:5166–5170

    PubMed  CAS  Google Scholar 

  • Sauer B, Henderson N (1989) Cre-stimulated recombination at loxP-containing DNA sequences placed into the mammalian genome. Nucleic Acids Res 17:147–161

    PubMed  CAS  Google Scholar 

  • Schaefer A, Im HI, Veno MT, Fowler CD, Min A, Intrator A, Kjems J, Kenny PJ, O’Carroll D, Greengard P (2010) Argonaute 2 in dopamine 2 receptor-expressing neurons regulates cocaine addiction. J Exp Med 207:1843–1851

    PubMed  CAS  Google Scholar 

  • Schlaepfer IR, Hoft NR, Collins AC, Corley RP, Hewitt JK, Hopfer CJ, Lessem JM, McQueen MB, Rhee SH, Ehringer MA (2008) The CHRNA5/A3/B4 gene cluster variability as an important determinant of early alcohol and tobacco initiation in young adults. Biol Psychiatry 63:1039–1046

    PubMed  CAS  Google Scholar 

  • Shen W, Flajolet M, Greengard P, Surmeier DJ (2008) Dichotomous dopaminergic control of striatal synaptic plasticity. Science 321:848–851

    PubMed  CAS  Google Scholar 

  • Sherva R, Wilhelmsen K, Pomerleau CS, Chasse SA, Rice JP, Snedecor SM, Bierut LJ, Neuman RJ, Pomerleau OF (2008) Association of a single nucleotide polymorphism in neuronal acetylcholine receptor subunit alpha 5 (CHRNA5) with smoking status and with ‘pleasurable buzz’ during early experimentation with smoking. Addiction 103:1544–1552

    PubMed  Google Scholar 

  • Sherva R, Kranzler HR, Yu Y, Logue MW, Poling J, Arias AJ, Anton RF, Oslin D, Farrer LA, Gelernter J (2010) Variation in nicotinic acetylcholine receptor genes is associated with multiple substance dependence phenotypes. Neuropsychopharmacology 35:1921–1931

    PubMed  CAS  Google Scholar 

  • Shizuya H, Birren B, Kim UJ, Mancino V, Slepak T, Tachiiri Y, Simon M (1992) Cloning and stable maintenance of 300-kilobase-pair fragments of human DNA in Escherichia coli using an F-factor-based vector. Proc Natl Acad Sci USA 89:8794–8797

    PubMed  CAS  Google Scholar 

  • Skarnes WC, Rosen B, West AP, Koutsourakis M, Bushell W, Iyer V, Mujica AO, Thomas M, Harrow J, Cox T, Jackson D, Severin J, Biggs P, Fu J, Nefedov M, de Jong PJ, Stewart AF, Bradley A (2011) A conditional knockout resource for the genome-wide study of mouse gene function. Nature 474:337–342

    PubMed  CAS  Google Scholar 

  • Sora I, Hall FS, Andrews AM, Itokawa M, Li XF, Wei HB, Wichems C, Lesch KP, Murphy DL, Uhl GR (2001) Molecular mechanisms of cocaine reward: combined dopamine and serotonin transporter knockouts eliminate cocaine place preference. Proc Natl Acad Sci USA 98:5300–5305

    PubMed  CAS  Google Scholar 

  • Sora I, Li B, Igari M, Hall FS, Ikeda K (2010) Transgenic mice in the study of drug addiction and the effects of psychostimulant drugs. Ann NY Acad Sci 1187:218–246

    PubMed  CAS  Google Scholar 

  • Sternberg N, Hamilton D, Austin S, Yarmolinsky M, Hoess R (1981) Site-specific recombination and its role in the life cycle of bacteriophage P1. Cold Spring Harb Symp Quant Biol 45(Pt 1):297–309

    PubMed  CAS  Google Scholar 

  • Steuber-Buchberger P, Wurst W, Kuhn R (2008) Simultaneous Cre-mediated conditional knockdown of two genes in mice. Genesis 46:144–151

    PubMed  CAS  Google Scholar 

  • Stuber GD, Hnasko TS, Britt JP, Edwards RH, Bonci A (2010) Dopaminergic terminals in the nucleus accumbens but not the dorsal striatum corelease glutamate. J Neurosci 30:8229–8233

    PubMed  CAS  Google Scholar 

  • Stuber GD, Sparta DR, Stamatakis AM, van Leeuwen WA, Hardjoprajitno JE, Cho S, Tye KM, Kempadoo KA, Zhang F, Deisseroth K, Bonci A (2011) Excitatory transmission from the amygdala to nucleus accumbens facilitates reward seeking. Nature 475:377–380

    PubMed  CAS  Google Scholar 

  • Sutherland HG, Kearns M, Morgan HD, Headley AP, Morris C, Martin DI, Whitelaw E (2000) Reactivation of heritably silenced gene expression in mice. Mamm Genome 11:347–355

    PubMed  CAS  Google Scholar 

  • Tapper AR, McKinney SL, Nashmi R, Schwarz J, Deshpande P, Labarca C, Whiteaker P, Marks MJ, Collins AC, Lester HA (2004) Nicotine activation of alpha4* receptors: sufficient for reward, tolerance, and sensitization. Science 306:1029–1032

    PubMed  CAS  Google Scholar 

  • Tate PH, Skarnes WC (2011) Bi-allelic gene targeting in mouse embryonic stem cells. Methods 53:331–338

    PubMed  CAS  Google Scholar 

  • Thomsen M, Han DD, Gu HH, Caine SB (2009) Lack of cocaine self-administration in mice expressing a cocaine-insensitive dopamine transporter. J Pharmacol Exp Ther 331:204–211

    PubMed  CAS  Google Scholar 

  • Thorgeirsson TE, Geller F, Sulem P, Rafnar T, Wiste A, Magnusson KP, Manolescu A, Thorleifsson G, Stefansson H, Ingason A, Stacey SN, Bergthorsson JT, Thorlacius S, Gudmundsson J, Jonsson T, Jakobsdottir M, Saemundsdottir J, Olafsdottir O, Gudmundsson LJ, Bjornsdottir G, Kristjansson K, Skuladottir H, Isaksson HJ, Gudbjartsson T, Jones GT, Mueller T, Gottsater A, Flex A, Aben KK, de Vegt F, Mulders PF, Isla D, Vidal MJ, Asin L, Saez B, Murillo L, Blondal T, Kolbeinsson H, Stefansson JG, Hansdottir I, Runarsdottir V, Pola R, Lindblad B, van Rij AM, Dieplinger B, Haltmayer M, Mayordomo JI, Kiemeney LA, Matthiasson SE, Oskarsson H, Tyrfingsson T, Gudbjartsson DF, Gulcher JR, Jonsson S, Thorsteinsdottir U, Kong A, Stefansson K (2008) A variant associated with nicotine dependence, lung cancer and peripheral arterial disease. Nature 452:638–642

    PubMed  CAS  Google Scholar 

  • Tilley MR, Gu HH (2008) Dopamine transporter inhibition is required for cocaine-induced stereotypy. Neuroreport 19:1137–1140

    PubMed  CAS  Google Scholar 

  • Tilley MR, O’Neill B, Han DD, Gu HH (2009) Cocaine does not produce reward in absence of dopamine transporter inhibition. Neuroreport 20:9–12

    PubMed  CAS  Google Scholar 

  • Trigo JM, Martin-Garcia E, Berrendero F, Robledo P, Maldonado R (2010) The endogenous opioid system: a common substrate in drug addiction. Drug Alcohol Depend 108:183–194

    PubMed  CAS  Google Scholar 

  • Tsai HC, Zhang F, Adamantidis A, Stuber GD, Bonci A, de Lecea L, Deisseroth K (2009) Phasic firing in dopaminergic neurons is sufficient for behavioral conditioning. Science 324:1080–1084

    PubMed  CAS  Google Scholar 

  • Tuesta L, Fowler CD, Kenny PJ (2011) Recent advances in understanding nicotinic receptor signaling mechanisms that regulate drug self-administration behavior. Biochem Pharmacol 82:984–995

    Google Scholar 

  • Valverde O, Mantamadiotis T, Torrecilla M, Ugedo L, Pineda J, Bleckmann S, Gass P, Kretz O, Mitchell JM, Schutz G, Maldonado R (2004) Modulation of anxiety-like behavior and morphine dependence in CREB-deficient mice. Neuropsychopharmacology 29:1122–1133

    PubMed  CAS  Google Scholar 

  • Vialou V, Robison AJ, Laplant QC, Covington HE 3rd, Dietz DM, Ohnishi YN, Mouzon E, Rush AJ 3rd, Watts EL, Wallace DL, Iniguez SD, Ohnishi YH, Steiner MA, Warren BL, Krishnan V, Bolanos CA, Neve RL, Ghose S, Berton O, Tamminga CA, Nestler EJ (2010) DeltaFosB in brain reward circuits mediates resilience to stress and antidepressant responses. Nat Neurosci 13:745–752

    PubMed  CAS  Google Scholar 

  • Wang JC, Grucza R, Cruchaga C, Hinrichs AL, Bertelsen S, Budde JP, Fox L, Goldstein E, Reyes O, Saccone N, Saccone S, Xuei X, Bucholz K, Kuperman S, Nurnberger J Jr, Rice JP, Schuckit M, Tischfield J, Hesselbrock V, Porjesz B, Edenberg HJ, Bierut LJ, Goate AM (2009) Genetic variation in the CHRNA5 gene affects mRNA levels and is associated with risk for alcohol dependence. Mol Psychiatry 14:501–510

    PubMed  CAS  Google Scholar 

  • Wang LP, Li F, Shen X, Tsien JZ (2010) Conditional knockout of NMDA receptors in dopamine neurons prevents nicotine-conditioned place preference. PLoS One 5:e8616

    PubMed  Google Scholar 

  • Wetterling T, Veltrup C, Driessen M, John U (1999) Drinking pattern and alcohol-related medical disorders. Alcohol Alcohol 34:330–336

    PubMed  CAS  Google Scholar 

  • Witten IB, Lin SC, Brodsky M, Prakash R, Diester I, Anikeeva P, Gradinaru V, Ramakrishnan C, Deisseroth K (2010) Cholinergic interneurons control local circuit activity and cocaine conditioning. Science 330:1677–1681

    PubMed  CAS  Google Scholar 

  • Wong DF, Kuwabara H, Schretlen DJ, Bonson KR, Zhou Y, Nandi A, Brasic JR, Kimes AS, Maris MA, Kumar A, Contoreggi C, Links J, Ernst M, Rousset O, Zukin S, Grace AA, Lee JS, Rohde C, Jasinski DR, Gjedde A, London ED (2006) Increased occupancy of dopamine receptors in human striatum during cue-elicited cocaine craving. Neuropsychopharmacology 31:2716–2727

    PubMed  CAS  Google Scholar 

  • Wu Y, Wang C, Sun H, LeRoith D, Yakar S (2009) High-efficient FLPo deleter mice in C57BL/6J background. PLoS One 4:e8054

    PubMed  Google Scholar 

  • Wu G, Sun Y, Qu W, Huang Y, Lu L, Li L, Shao W (2011) Application of GFAT as a novel selection marker to mediate gene expression. PLoS One 6:e17082

    PubMed  CAS  Google Scholar 

  • Yamamoto M, Wada N, Kitabatake Y, Watanabe D, Anzai M, Yokoyama M, Teranishi Y, Nakanishi S (2003) Reversible suppression of glutamatergic neurotransmission of cerebellar granule cells in vivo by genetically manipulated expression of tetanus neurotoxin light chain. J Neurosci 23:6759–6767

    PubMed  CAS  Google Scholar 

  • Ye H, Daoud-El Baba M, Peng RW, Fussenegger M (2011) A synthetic optogenetic transcription device enhances blood-glucose homeostasis in mice. Science 332:1565–1568

    PubMed  CAS  Google Scholar 

  • Yizhar O, Fenno LE, Davidson TJ, Mogri M, Deisseroth K (2011) Optogenetics in neural systems. Neuron 71:9–34

    PubMed  CAS  Google Scholar 

  • Zhao ZQ, Scott M, Chiechio S, Wang JS, Renner KJ, Gereau RWt, Johnson RL, Deneris ES, Chen ZF (2006) Lmx1b is required for maintenance of central serotonergic neurons and mice lacking central serotonergic system exhibit normal locomotor activity. J Neurosci 26:12781–12788

    PubMed  CAS  Google Scholar 

  • Zhao ZQ, Chiechio S, Sun YG, Zhang KH, Zhao CS, Scott M, Johnson RL, Deneris ES, Renner KJ, Gereau RWt, Chen ZF (2007a) Mice lacking central serotonergic neurons show enhanced inflammatory pain and an impaired analgesic response to antidepressant drugs. J Neurosci 27:6045–6053

    PubMed  CAS  Google Scholar 

  • Zhao ZQ, Gao YJ, Sun YG, Zhao CS, Gereau RWt, Chen ZF (2007b) Central serotonergic neurons are differentially required for opioid analgesia but not for morphine tolerance or morphine reward. Proc Natl Acad Sci USA 104:14519–14524

    PubMed  CAS  Google Scholar 

  • Zhao-Shea R, Cohen BN, Just H, McClure-Begley T, Whiteaker P, Grady SR, Salminen O, Gardner PD, Lester HA, Tapper AR (2010) Dopamine D2-receptor activation elicits akinesia, rigidity, catalepsy, and tremor in mice expressing hypersensitive {alpha}4 nicotinic receptors via a cholinergic-dependent mechanism. FASEB J 24:49–57

    PubMed  Google Scholar 

  • Zhao-Shea R, Liu L, Soll LG, Improgo MR, Meyers EE, McIntosh JM, Grady SR, Marks MJ, Gardner PD, Tapper AR (2011) Nicotine-mediated activation of dopaminergic neurons in distinct regions of the ventral tegmental area. Neuropsychopharmacology 36:1021–1032

    PubMed  CAS  Google Scholar 

  • Zhou Y, Litvin Y, Piras AP, Pfaff DW, Kreek MJ (2011) Persistent increase in hypothalamic arginine vasopressin gene expression during protracted withdrawal from chronic escalating-dose cocaine in rodents. Neuropsychopharmacology 36:2062–2075

    Google Scholar 

  • Zweifel LS, Argilli E, Bonci A, Palmiter RD (2008) Role of NMDA receptors in dopamine neurons for plasticity and addictive behaviors. Neuron 59:486–496

    PubMed  CAS  Google Scholar 

Download references

Acknowledgment

Supported by the National Institute on Drug Abuse (DA020686 to PJK; DA026693 to CDF).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Paul J. Kenny.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Fowler, C.D., Kenny, P.J. Utility of genetically modified mice for understanding the neurobiology of substance use disorders. Hum Genet 131, 941–957 (2012). https://doi.org/10.1007/s00439-011-1129-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00439-011-1129-z

Keywords

Navigation