Skip to main content
Log in

Genetic background influences the effects of withdrawal from chronic nicotine on learning and high-affinity nicotinic acetylcholine receptor binding in the dorsal and ventral hippocampus

  • Original Investigation
  • Published:
Psychopharmacology Aims and scope Submit manuscript

Abstract

Rationale

The effects of nicotine on cognitive processes may play an important role in nicotine addiction. Nicotine withdrawal impairs hippocampus-dependent learning and genetic factors influence this effect. However, the neural changes that contribute to these impairments are unknown. Chronic nicotine upregulates hippocampal nicotinic acetycholine receptors (nAChRs), which may contribute to cognitive deficits when nicotine administration ceases. If nAChR upregulation underlies withdrawal deficits in learning, then strains of mice exhibiting withdrawal deficits in hippocampus-dependent learning should also show upregulation of hippocampal nAChRs.

Objectives

Here, we examined the effects of nicotine withdrawal on fear conditioning and [3H]epibatidine binding in the dorsal and ventral hippocampus in two inbred mouse strains and their F1 hybrids.

Methods

Male C57BL/6NTac, 129S6/SvEvTac, and B6129SF1/Tac mice were administered chronic nicotine (18 mg/kg/day) for 12 days through osmotic pumps and then were trained and tested in fear conditioning 24 h after cessation of nicotine treatment.

Results

Nicotine withdrawal impaired hippocampus-dependent contextual conditioning in C57BL/6NTac mice but not 129S6/SvEvTac or B6129SF1/Tac mice; no changes were observed in hippocampus-independent cued fear conditioning. Upregulated [3H]epibatidine binding was found in the dorsal, but not ventral, hippocampus of C57BL/6NTac mice and in the ventral hippocampus of B6129SF1/Tac mice after chronic nicotine.

Conclusions

Upregulation of high-affinity binding sites in the dorsal hippocampus of C57BL/6NTac mice, the only strain that exhibited nAChR upregulation in this region and withdrawal deficits in contextual conditioning, suggests that upregulation of high-affinity binding sites in the dorsal hippocampus mediates, in part, nicotine withdrawal deficits in contextual conditioning and genetic background modulates these effects.

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.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  • Abdulla FA, Bradbury E, Calaminici MR, Lippiello PM, Wonnacott S, Gray JA, Sinden JD (1996) Relationship between up-regulation of nicotine binding sites in rat brain and delayed cognitive enhancement observed after chronic or acute nicotinic receptor stimulation. Psychopharmacol (Berl) 124:323–331

    Article  CAS  Google Scholar 

  • Benowitz NL (2008) Clinical pharmacology of nicotine: implications for understanding, preventing, and treating tobacco addiction. Clin Pharmacol Ther 83:531–541

    Article  PubMed  CAS  Google Scholar 

  • Bothe GWM, Bolivar VJ, Vedder MJ, Geistfeld JG (2004) Genetic and behavioral differences among five inbred mouse strains commonly used in the production of transgenic and knockout mice. Genes Brain Behav 3:149–157

    Article  PubMed  CAS  Google Scholar 

  • Bothe GWM, Bolivar VJ, Vedder MJ, Geistfeld JG (2005) Behavioral differences among fourteen inbred mouse strains commonly used as disease models. Comp Med 55:326–334

    PubMed  CAS  Google Scholar 

  • Bryant CD, Zhang NN, Sokoloff G, Fanselow MS, Ennes HS, Palmer AA, McRoberts JA (2008) Behavioral differences among C57BL/6 substrains: implications for transgenic and knockout studies. J Neurogenet 22:315–331

    Article  PubMed  CAS  Google Scholar 

  • CDC (2011) CDC health disparities and inequalities report—United States, 2011. MMWR 60:109–112

    Google Scholar 

  • Clapcote SJ, Roder JC (2004) Survey of embryonic stem cell line source strains in the water maze reveals superior reversal learning of 129S6/SvEvTac mice. Behav Brain Res 152:35–48

    PubMed  Google Scholar 

  • Collins AC, Miner LL, Marks MJ (1988) Genetic influences on acute responses to nicotine and nicotine tolerance in the mouse. Pharmacol Biochem Behav 30:269–278

    Article  PubMed  CAS  Google Scholar 

  • Committee on Standardized Genetic Nomenclature for Mice (1989) Rules for nomenclature of inbred strains. In: Lyon MF, Searle AG (eds) Genetic variants and strains of the laboratory mouse. Oxford University Press, New York, pp 632–635

    Google Scholar 

  • Crawley JN, Belknap JK, Collins A, Crabbe JC, Frankel W, Henderson N, Hitzemann RJ, Maxson SC, Miner LL, Silva AJ, Wehner JM, Wynshaw-Boris A, Paylor R (1997) Behavioral phenotypes of inbred mouse strains: implications and recommendations for molecular studies. Psychopharmacol (Berl) 132:107–124

    Article  CAS  Google Scholar 

  • Damaj MI, Kao W, Martin BR (2003) Characterization of spontaneous and precipitated nicotine withdrawal in the mouse. J Pharmacol Exp Ther 307:526–534

    Article  PubMed  CAS  Google Scholar 

  • Damaj MI, Fonck C, Marks MJ, Deshpande P, Labarca C, Lester HA, Collins AC, Martin BR (2007) Genetic approaches identify differential roles for alpha4beta2* nicotinic receptors in acute models of antinociception in mice. J Pharmacol Exp Ther 321:1161–1169

    Article  PubMed  CAS  Google Scholar 

  • Davis JA, Gould TJ (2009) Hippocampal nAChRs mediate nicotine withdrawal-related learning deficits. Eur Neuropsychopharmacol 19:551–561

    Article  PubMed  CAS  Google Scholar 

  • Davis JA, James JR, Siegel SJ, Gould TJ (2005) Withdrawal from chronic nicotine administration impairs contextual fear conditioning in C57BL/6 mice. J Neurosci 25:8708–8713

    Article  PubMed  CAS  Google Scholar 

  • Davis JA, Kenney JW, Gould TJ (2007) Hippocampal alpha4beta2 nicotinic acetylcholine receptor involvement in the enhancing effect of acute nicotine on contextual fear conditioning. J Neurosci 27:10870–10877

    Article  PubMed  CAS  Google Scholar 

  • Evans DE, Drobes DJ (2009) Nicotine self-medication of cognitive-attentional processing. Addict Biol 14:32–42

    Article  PubMed  CAS  Google Scholar 

  • Fanselow MS, Dong HW (2010) Are the dorsal and ventral hippocampus functionally distinct structures? Neuron 65:7–19

    Article  PubMed  CAS  Google Scholar 

  • Gould TJ, Wehner JM (1999a) Genetic influences on latent inhibition. Behav Neurosci 113:1291–1296

    Article  PubMed  CAS  Google Scholar 

  • Gould TJ, Wehner JM (1999b) Nicotine enhancement of contextual fear conditioning. Behav Brain Res 102:31–39

    Article  PubMed  CAS  Google Scholar 

  • Gould TJ, Feiro O, Moore D (2004) Nicotine enhances trace cued fear conditioning but not delay cued fear conditioning in C57BL/6 mice. Behav Brain Res 155:167–173

    Article  PubMed  CAS  Google Scholar 

  • Gould TJ, Portugal GS, André JM, Tadman MP, Marks MJ, Kenney JW, Yildirim E, Adoff M (2012) The duration of nicotine withdrawal-associated deficits in contextual fear conditioning parallels changes in hippocampal high affinity nicotinic acetylcholine receptor upregulation. Neuropharmacology 62:2118–2125

    Article  PubMed  CAS  Google Scholar 

  • Govind AP, Vezina P, Green WN (2009) Nicotine-induced upregulation of nicotinic receptors: underlying mechanisms and relevance to nicotine addiction. Biochem Pharmacol 78:756–765

    Article  PubMed  CAS  Google Scholar 

  • Govind AP, Walsh H, Green WN (2012) Nicotine-induced upregulation of native neuronal nicotinic receptors is caused by multiple mechanisms. J Neurosci 32:2227–2238

    Article  PubMed  CAS  Google Scholar 

  • Gresack JE, Schafe GE, Orr PT, Frick KM (2009) Sex differences in contextual fear conditioning are associated with differential ventral hippocampal extracellular signal-regulated kinase activation. Neuroscience 159:451–467

    Article  PubMed  CAS  Google Scholar 

  • Jacobsen LK, Mencl WE, Constable RT, Westerveld M, Pugh KR (2007) Impact of smoking abstinence on working memory neurocircuitry in adolescent daily tobacco smokers. Psychopharmacol (Berl) 193:557–566

    Article  CAS  Google Scholar 

  • Kenney JW, Raybuck JD, Gould TJ (2012) Nicotinic receptors in the dorsal and ventral hippocampus differentially modulate contextual fear conditioning. Hippocampus 22(8):1681–1690

    Article  PubMed  CAS  Google Scholar 

  • Kenny PJ, Markou A (2001) Neurobiology of the nicotine withdrawal syndrome. Pharmacol Biochem Behav 70:531–549

    Article  PubMed  CAS  Google Scholar 

  • Kiselycznyk C, Holmes A (2011) All (C57BL/6) mice are not created equal. Front Neurosci 5:10–10

    Article  PubMed  Google Scholar 

  • Logue SF, Paylor R, Wehner JM (1997) Hippocampal lesions cause learning deficits in inbred mice in the Morris water maze and conditioned-fear task. Behav Neurosci 111:104–113

    Article  PubMed  CAS  Google Scholar 

  • Marks MJ, Burch JB, Collins AC (1983a) Effects of chronic nicotine infusion on tolerance development and nicotinic receptors. J Pharmacol Exp Ther 226:817–825

    PubMed  CAS  Google Scholar 

  • Marks MJ, Burch JB, Collins AC (1983b) Genetics of nicotine response in four inbred strains of mice. J Pharmacol Exp Ther 226:291–302

    PubMed  CAS  Google Scholar 

  • Marks MJ, Stitzel JA, Collins AC (1985) Time course study of the effects of chronic nicotine infusion on drug response and brain receptors. J Pharmacol Exp Ther 235:619–628

    PubMed  CAS  Google Scholar 

  • Marks MJ, Campbell SM, Romm E, Collins AC (1991) Genotype influences the development of tolerance to nicotine in the mouse. J Pharmacol Exp Ther 259:392–402

    PubMed  CAS  Google Scholar 

  • Moser MB, Moser EI, Forrest E, Andersen P, Morris RG (1995) Spatial learning with a minislab in the dorsal hippocampus. Proc Natl Acad Sci U S A 92:9697–9701

    Article  PubMed  CAS  Google Scholar 

  • Nie T, Abel T (2001) Fear conditioning in inbred mouse strains: an analysis of the time course of memory. Behav Neurosci 115:951–956

    Article  PubMed  CAS  Google Scholar 

  • Owen EH, Logue SF, Rasmussen DL, Wehner JM (1997) Assessment of learning by the Morris water task and fear conditioning in inbred mouse strains and F1 hybrids: implications of genetic background for single gene mutations and quantitative trait loci analyses. Neuroscience 80:1087–1099

    Article  PubMed  CAS  Google Scholar 

  • Patterson F, Jepson C, Loughead J, Perkins K, Strasser AA, Siegel S, Frey J, Gur R, Lerman C (2010) Working memory deficits predict short-term smoking resumption following brief abstinence. Drug Alcohol Depend 106:61–64

    Article  PubMed  Google Scholar 

  • Pergadia ML, Heath AC, Martin NG, Madden PAF (2006) Genetic analyses of DSM-IV nicotine withdrawal in adult twins. Psychol Med 36:963–972

    Article  PubMed  Google Scholar 

  • Perry DC, Xiao Y, Nguyen HN, Musachio JL, Dávila-García MI, Kellar KJ (2002) Measuring nicotinic receptors with characteristics of alpha4beta2, alpha3beta2 and alpha3beta4 subtypes in rat tissues by autoradiography. J Neurochem 82:468–481

    Article  PubMed  CAS  Google Scholar 

  • Phillips RG, LeDoux JE (1992) Differential contribution of amygdala and hippocampus to cued and contextual fear conditioning. Behav Neurosci 106:274–285

    Article  PubMed  CAS  Google Scholar 

  • Piasecki TM, Fiore MC, Baker TB (1998) Profiles in discouragement: two studies of variability in the time course of smoking withdrawal symptoms. J Abnorm Psychol 107:238–251

    Article  PubMed  CAS  Google Scholar 

  • Piasecki TM, Niaura R, Shadel WG, Abrams D, Goldstein M, Fiore MC, Baker TB (2000) Smoking withdrawal dynamics in unaided quitters. J Abnorm Psychol 109:74–86

    Article  PubMed  CAS  Google Scholar 

  • Portugal GS, Gould TJ (2008) Genetic variability in nicotinic acetylcholine receptors and nicotine addiction: converging evidence from human and animal research. Behav Brain Res 193:1–16

    Article  PubMed  CAS  Google Scholar 

  • Portugal GS, Gould TJ (2009) Nicotine withdrawal disrupts new contextual learning. Pharmacol Biochem Behav 92:117–123

    Article  PubMed  CAS  Google Scholar 

  • Portugal GS, Kenney JW, Gould TJ (2008) Beta2 subunit containing acetylcholine receptors mediate nicotine withdrawal deficits in the acquisition of contextual fear conditioning. Neurobiol Learn Mem 89:106–113

    Article  PubMed  CAS  Google Scholar 

  • Portugal GS, Wilkinson DS, Kenney JW, Sullivan C, Gould TJ (2012a) Strain-dependent effects of acute, chronic, and withdrawal from chronic nicotine on fear conditioning. Behav Genet 42:133–150

    Article  PubMed  Google Scholar 

  • Portugal GS, Wilkinson DS, Turner JR, Blendy J, Gould TJ (2012b) Developmental effects of acute, chronic, and withdrawal from chronic nicotine on fear conditioning. Neurobiol Learn Mem 97:482–494

    Article  PubMed  CAS  Google Scholar 

  • Raybuck JD, Gould TJ (2010) The role of nicotinic acetylcholine receptors in the medial prefrontal cortex and hippocampus in trace fear conditioning. Neurobiol Learn Mem 94:353–363

    Article  PubMed  CAS  Google Scholar 

  • SAMHSA (2010) Results from the 2010 National Survey on Drug Use and Health: summary of national findings. NSDUH Series H-41, HHS Publication No (SMA) 11-4658 Rockville, MD

  • Schwartz RD, Kellar KJ (1983) Nicotinic cholinergic receptor binding sites in the brain: regulation in vivo. Science 220:214–216

    Article  PubMed  CAS  Google Scholar 

  • Stiedl O, Radulovic J, Lohmann R, Birkenfeld K, Palve M, Kammermeier J, Sananbenesi F, Spiess J (1999) Strain and substrain differences in context- and tone-dependent fear conditioning of inbred mice. Behav Brain Res 104:1–12

    Article  PubMed  CAS  Google Scholar 

  • True WR, Heath AC, Scherrer JF, Waterman B, Goldberg J, Lin N, Eisen SA, Lyons MJ, Tsuang MT (1997) Genetic and environmental contributions to smoking. Addiction 92:1277–1287

    Article  PubMed  CAS  Google Scholar 

  • Turner JR, Castellano LM, Blendy JA (2011a) Parallel anxiolytic-like effects and upregulation of neuronal nicotinic acetylcholine receptors following chronic nicotine and varenicline. Nicotine Tob Res 13:41–46

    Article  PubMed  CAS  Google Scholar 

  • Turner JR, Ortinski PI, Sherrard RM, Kellar KJ (2011b) Cerebellar nicotinic cholinergic receptors are intrinsic to the cerebellum: implications for diverse functional roles. Cerebellum 10:748–757

    Article  PubMed  CAS  Google Scholar 

  • Wehner JM, Radcliffe RA, Bowers BJ (2001) Quantitative genetics and mouse behavior. Annu Rev Neurosci 24:845–867

    Article  PubMed  CAS  Google Scholar 

  • Wolff M, Savova M, Malleret G, Segu L, Buhot MC (2002) Differential learning abilities of 129T2/Sv and C57BL/6J mice as assessed in three water maze protocols. Behav Brain Res 136:463–474

    Article  PubMed  Google Scholar 

  • Xiao Y, Kellar KJ (2004) The comparative pharmacology and up-regulation of rat neuronal nicotinic receptor subtype binding sites stably expressed in transfected mammalian cells. J Pharmacol Exp Ther 310:98–107

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

The authors would like to acknowledge grant support from the National Institute on Drug Abuse (NIDA, DA024787, DA017949, TJG; DA026236, JRT) and the National Cancer Institute (CA143187, JAB). DSW was supported by a NIDA diversity supplement (DA024787-01A1S1). All procedures were in accordance with the NIH Guidelines for the Care and Use of Laboratory Animals and US laws.

Conflict of interest

The authors report no conflicts of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Thomas J. Gould.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wilkinson, D.S., Turner, J.R., Blendy, J.A. et al. Genetic background influences the effects of withdrawal from chronic nicotine on learning and high-affinity nicotinic acetylcholine receptor binding in the dorsal and ventral hippocampus. Psychopharmacology 225, 201–208 (2013). https://doi.org/10.1007/s00213-012-2808-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00213-012-2808-8

Keywords

Navigation