Skip to main content
Top
Published in: Brain Structure and Function 4/2016

01-05-2016 | Original Article

Impact of prenatal nicotine on the structure of midbrain dopamine regions in the rat

Authors: Natalia Omelchenko, Priya Roy, Judith Joyce Balcita-Pedicino, Samuel Poloyac, Susan R. Sesack

Published in: Brain Structure and Function | Issue 4/2016

Login to get access

Abstract

In utero exposure of rats to nicotine (NIC) provides a useful animal model for studying the impact of smoking during pregnancy on human offspring. Certain sequelae of prenatal NIC exposure suggest an impact on the development of the midbrain dopamine (DA) system, which receives a robust cholinergic innervation from the mesopontine tegmentum. We therefore investigated whether prenatal NIC induced structural changes in cells and synapses within the midbrain that persisted into adulthood. Osmotic minipumps delivering either sodium bitartrate (vehicle; VEH) or NIC bitartrate at 2 mg/kg/day were implanted into nine timed-pregnant dams at E4. At birth, rat pups were culled to litters of six males each, and the litters were cross-fostered. Plasma levels of NIC and cotinine from killed pups provided evidence of NIC exposure in utero. Pups separated from dams at weaning showed a trend toward reduced locomotor activity at this time point but not when tested again in adulthood. Adult rats were killed for anatomical studies. Estimates of brain size and volume did not vary with NIC treatment. Midbrain sections stained for Nissl or by immunoperoxidase for tyrosine hydroxylase and analyzed using unbiased stereology revealed no changes in volume or cell number in the substantia nigra compacta or ventral tegmental area as a result of NIC exposure. Within the ventral tegmental area, electron microscopic physical disector analysis showed no significant differences in the number of axon terminals or the number of asymmetric (putative excitatory) or symmetric (putative inhibitory) synapses. Although too infrequent to estimate by unbiased stereology, no obvious difference in the proportion of cholinergic axons was noted in NIC- versus VEH-treated animals. These data suggest that activation of nicotinic receptors during prenatal development induces no significant modifications in the structure of cells in the ventral midbrain when assessed in adulthood.
Literature
go back to reference Abdel-Rahman A, Dechkovskaia AM, Sutton JM, Chen WC, Guan X, Khan WA, Abou-Donia MB (2005) Maternal exposure of rats to nicotine via infusion during gestation produces neurobehavioral deficits and elevated expression of glial fibrillary acidic protein in the cerebellum and CA1 subfield in the offspring at puberty. Toxicology 209:245–261PubMedCrossRef Abdel-Rahman A, Dechkovskaia AM, Sutton JM, Chen WC, Guan X, Khan WA, Abou-Donia MB (2005) Maternal exposure of rats to nicotine via infusion during gestation produces neurobehavioral deficits and elevated expression of glial fibrillary acidic protein in the cerebellum and CA1 subfield in the offspring at puberty. Toxicology 209:245–261PubMedCrossRef
go back to reference Abreu-Villaca Y, Seidler FJ, Tate CA, Slotkin TA (2003) Nicotine is a neurotoxin in the adolescent brain: critical periods, patterns of exposure, regional selectivity, and dose thresholds for macromolecular alterations. Brain Res 979:114–128PubMedCrossRef Abreu-Villaca Y, Seidler FJ, Tate CA, Slotkin TA (2003) Nicotine is a neurotoxin in the adolescent brain: critical periods, patterns of exposure, regional selectivity, and dose thresholds for macromolecular alterations. Brain Res 979:114–128PubMedCrossRef
go back to reference Abreu-Villaca Y, Seidler FJ, Slotkin TA (2004a) Does prenatal nicotine exposure sensitize the brain to nicotine-induced neurotoxicity in adolescence? Neuropsychopharmacology 29:1440–1450PubMedCrossRef Abreu-Villaca Y, Seidler FJ, Slotkin TA (2004a) Does prenatal nicotine exposure sensitize the brain to nicotine-induced neurotoxicity in adolescence? Neuropsychopharmacology 29:1440–1450PubMedCrossRef
go back to reference Abreu-Villaca Y, Seidler FJ, Tate CA, Cousins MM, Slotkin TA (2004b) Prenatal nicotine exposure alters the response to nicotine administration in adolescence: effects on cholinergic systems during exposure and withdrawal. Neuropsychopharmacology 29:879–890PubMedCrossRef Abreu-Villaca Y, Seidler FJ, Tate CA, Cousins MM, Slotkin TA (2004b) Prenatal nicotine exposure alters the response to nicotine administration in adolescence: effects on cholinergic systems during exposure and withdrawal. Neuropsychopharmacology 29:879–890PubMedCrossRef
go back to reference Adriani W, Macri S, Pacifici R, Laviola G (2002) Peculiar vulnerability to nicotine oral self-administration in mice during early adolescence. Neuropsychopharmacology 27:212–224PubMedCrossRef Adriani W, Macri S, Pacifici R, Laviola G (2002) Peculiar vulnerability to nicotine oral self-administration in mice during early adolescence. Neuropsychopharmacology 27:212–224PubMedCrossRef
go back to reference Adriani W, Spijker S, Deroche-Gamonet V, Laviola G, Le Moal M, Smit AB, Piazza PV (2003) Evidence for enhanced neurobehavioral vulnerability to nicotine during periadolescence in rats. J Neurosci 23:4712–4716PubMed Adriani W, Spijker S, Deroche-Gamonet V, Laviola G, Le Moal M, Smit AB, Piazza PV (2003) Evidence for enhanced neurobehavioral vulnerability to nicotine during periadolescence in rats. J Neurosci 23:4712–4716PubMed
go back to reference Ajarem JS, Ahmad M (1998) Prenatal nicotine exposure modifies behavior of mice through early development. Pharmacol Biochem Behav 59:313–318PubMedCrossRef Ajarem JS, Ahmad M (1998) Prenatal nicotine exposure modifies behavior of mice through early development. Pharmacol Biochem Behav 59:313–318PubMedCrossRef
go back to reference Andres RL, Day MC (2000) Perinatal complications associated with maternal tobacco use. Semin Neonatol 5:231–241PubMedCrossRef Andres RL, Day MC (2000) Perinatal complications associated with maternal tobacco use. Semin Neonatol 5:231–241PubMedCrossRef
go back to reference Atluri P, Fleck MW, Shen Q, Mah SJ, Stadfelt D, Barnes W, Goderie SK, Temple S, Schneider AS (2001) Functional nicotinic acetylcholine receptor expression in stem and progenitor cells of the early embryonic mouse cerebral cortex. Dev Biol 240:143–156PubMedCrossRef Atluri P, Fleck MW, Shen Q, Mah SJ, Stadfelt D, Barnes W, Goderie SK, Temple S, Schneider AS (2001) Functional nicotinic acetylcholine receptor expression in stem and progenitor cells of the early embryonic mouse cerebral cortex. Dev Biol 240:143–156PubMedCrossRef
go back to reference Audesirk T, Cabell L (1999) Nanomolar concentrations of nicotine and cotinine alter the development of cultured hippocampal neurons via non-acetylcholine receptor-mediated mechanisms. Neurotoxicology 20:639–646PubMed Audesirk T, Cabell L (1999) Nanomolar concentrations of nicotine and cotinine alter the development of cultured hippocampal neurons via non-acetylcholine receptor-mediated mechanisms. Neurotoxicology 20:639–646PubMed
go back to reference Bardy AH, Seppala T, Lillsunde P, Kataja JM, Koskela P, Pikkarainen J, Hiilesmaa VK (1993) Objectively measured tobacco exposure during pregnancy: neonatal effects and relation to maternal smoking. Br J Obstet Gynaecol 100:721–726PubMedCrossRef Bardy AH, Seppala T, Lillsunde P, Kataja JM, Koskela P, Pikkarainen J, Hiilesmaa VK (1993) Objectively measured tobacco exposure during pregnancy: neonatal effects and relation to maternal smoking. Br J Obstet Gynaecol 100:721–726PubMedCrossRef
go back to reference Bayer SA, Altman J (2007) The human brain during the early first trimester. Taylor & Francis, USACrossRef Bayer SA, Altman J (2007) The human brain during the early first trimester. Taylor & Francis, USACrossRef
go back to reference Bayer VE, Pickel VM (1990) Ultrastructural localization of tyrosine hydroxylase in the rat ventral tegmental area: relationship between immunolabeling density and neuronal associations. J Neurosci 10:2996–3013PubMed Bayer VE, Pickel VM (1990) Ultrastructural localization of tyrosine hydroxylase in the rat ventral tegmental area: relationship between immunolabeling density and neuronal associations. J Neurosci 10:2996–3013PubMed
go back to reference Bayer SA, Altman J, Russo RJ, Zhang X (1993) Timetables of neurogenesis in the human brain based on experimentally determined patterns in the rat. Neurotoxicology 14:83–144PubMed Bayer SA, Altman J, Russo RJ, Zhang X (1993) Timetables of neurogenesis in the human brain based on experimentally determined patterns in the rat. Neurotoxicology 14:83–144PubMed
go back to reference Benowitz NL (1996a) Biomarkers of cigarette smoking. The FTC cigarette test method for determining tar, nicotine, and carbon monoxide yields of U.S. cigarettes. U.S. Department of Health and Human Services, Public Health Service, National Institutes of Health, Bethesda, pp 93–111 Benowitz NL (1996a) Biomarkers of cigarette smoking. The FTC cigarette test method for determining tar, nicotine, and carbon monoxide yields of U.S. cigarettes. U.S. Department of Health and Human Services, Public Health Service, National Institutes of Health, Bethesda, pp 93–111
go back to reference Benowitz NL (1996b) Pharmacology of nicotine: addiction and therapeutics. Annu Rev Pharmacol Toxicol 36:597–613PubMedCrossRef Benowitz NL (1996b) Pharmacology of nicotine: addiction and therapeutics. Annu Rev Pharmacol Toxicol 36:597–613PubMedCrossRef
go back to reference Berger F, Gage FH, Vijayaraghavan S (1998) Nicotinic receptor-induced apoptotic cell death of hippocampal progenitor cells. J Neurosci 18:6871–6881PubMed Berger F, Gage FH, Vijayaraghavan S (1998) Nicotinic receptor-induced apoptotic cell death of hippocampal progenitor cells. J Neurosci 18:6871–6881PubMed
go back to reference Buchmann AF, Zohsel K, Blomeyer D, Hohm E, Hohmann S, Jennen-Steinmetz C, Treutlein J, Becker K, Banaschewski T, Schmidt MH, Esser G, Brandeis D, Poustka L, Zimmermann US, Laucht M (2014) Interaction between prenatal stress and dopamine D4 receptor genotype in predicting aggression and cortisol levels in young adults. Psychopharmacology 231:3089–3097PubMedCrossRef Buchmann AF, Zohsel K, Blomeyer D, Hohm E, Hohmann S, Jennen-Steinmetz C, Treutlein J, Becker K, Banaschewski T, Schmidt MH, Esser G, Brandeis D, Poustka L, Zimmermann US, Laucht M (2014) Interaction between prenatal stress and dopamine D4 receptor genotype in predicting aggression and cortisol levels in young adults. Psychopharmacology 231:3089–3097PubMedCrossRef
go back to reference Buka SL, Shenassa ED, Niaura R (2003) Elevated risk of tobacco dependence among offspring of mothers who smoked during pregnancy: a 30-year prospective study. Am J Psychiatry 160:1978–1984PubMedCrossRef Buka SL, Shenassa ED, Niaura R (2003) Elevated risk of tobacco dependence among offspring of mothers who smoked during pregnancy: a 30-year prospective study. Am J Psychiatry 160:1978–1984PubMedCrossRef
go back to reference Buznikov GA, Shmukler YB, Lauder JM (1996) From oocyte to neuron: do neurotransmitters function in the same way throughout development? Cell Mol Neurobiol 16:537–559PubMedCrossRef Buznikov GA, Shmukler YB, Lauder JM (1996) From oocyte to neuron: do neurotransmitters function in the same way throughout development? Cell Mol Neurobiol 16:537–559PubMedCrossRef
go back to reference Calverley RK, Bedi KS, Jones DG (1988) Estimation of the numerical density of synapses in rat neocortex. Comparison of the ‘disector’ with an ‘unfolding’ method. J Neurosci Methods 23:195–205PubMedCrossRef Calverley RK, Bedi KS, Jones DG (1988) Estimation of the numerical density of synapses in rat neocortex. Comparison of the ‘disector’ with an ‘unfolding’ method. J Neurosci Methods 23:195–205PubMedCrossRef
go back to reference Carr DB, Sesack SR (2000) Projections from the rat prefrontal cortex to the ventral tegmental area: target specificity in the synaptic associations with mesoaccumbens and mesocortical neurons. J Neurosci 20:3864–3873PubMed Carr DB, Sesack SR (2000) Projections from the rat prefrontal cortex to the ventral tegmental area: target specificity in the synaptic associations with mesoaccumbens and mesocortical neurons. J Neurosci 20:3864–3873PubMed
go back to reference Chen WJ, Edwards RB (2003) Prenatal nicotine exposure does not cause Purkinje cell loss in the developing rat cerebellar vermis. Neurotoxicol Teratol 25:633–637PubMedCrossRef Chen WJ, Edwards RB (2003) Prenatal nicotine exposure does not cause Purkinje cell loss in the developing rat cerebellar vermis. Neurotoxicol Teratol 25:633–637PubMedCrossRef
go back to reference Chen WJ, Parnell SE, West JR (1999) Effects of alcohol and nicotine on developing olfactory bulb: loss of mitral cells and alterations in neurotransmitter levels. Alcohol Clin Exp Res 23:18–25PubMed Chen WJ, Parnell SE, West JR (1999) Effects of alcohol and nicotine on developing olfactory bulb: loss of mitral cells and alterations in neurotransmitter levels. Alcohol Clin Exp Res 23:18–25PubMed
go back to reference Chen WJ, Edwards RB, Romero RD, Parnell SE, Monk RJ (2003) Long-term nicotine exposure reduces Purkinje cell number in the adult rat cerebellar vermis. Neurotoxicol Teratol 25:329–334PubMedCrossRef Chen WJ, Edwards RB, Romero RD, Parnell SE, Monk RJ (2003) Long-term nicotine exposure reduces Purkinje cell number in the adult rat cerebellar vermis. Neurotoxicol Teratol 25:329–334PubMedCrossRef
go back to reference Choy KH, de Visser YP, van den Buuse M (2009) The effect of ‘two hit’ neonatal and young-adult stress on dopaminergic modulation of prepulse inhibition and dopamine receptor density. Br J Pharmacol 156:388–396PubMedPubMedCentralCrossRef Choy KH, de Visser YP, van den Buuse M (2009) The effect of ‘two hit’ neonatal and young-adult stress on dopaminergic modulation of prepulse inhibition and dopamine receptor density. Br J Pharmacol 156:388–396PubMedPubMedCentralCrossRef
go back to reference Cornelius MD, Leech SL, Goldschmidt L, Day NL (2000) Prenatal tobacco exposure: is it a risk factor for early tobacco experimentation? Nicotine Tob Res 2:45–52PubMedCrossRef Cornelius MD, Leech SL, Goldschmidt L, Day NL (2000) Prenatal tobacco exposure: is it a risk factor for early tobacco experimentation? Nicotine Tob Res 2:45–52PubMedCrossRef
go back to reference Crooks PA, Dwoskin LP (1997) Contribution of CNS nicotine metabolites to the neuropharmacological effects of nicotine and tobacco smoking. Biochem Pharmacol 54:743–753PubMedCrossRef Crooks PA, Dwoskin LP (1997) Contribution of CNS nicotine metabolites to the neuropharmacological effects of nicotine and tobacco smoking. Biochem Pharmacol 54:743–753PubMedCrossRef
go back to reference Day M, Wang Z, Ding J, An X, Ingham CA, Shering AF, Wokosin D, Ilijic E, Sun Z, Sampson AR, Mugnaini E, Deutch AY, Sesack SR, Arbuthnott GW, Surmeier DJ (2006) Selective elimination of glutamatergic synapses on striatopallidal neurons in Parkinson disease models. Nat Neurosci 9:251–259PubMedCrossRef Day M, Wang Z, Ding J, An X, Ingham CA, Shering AF, Wokosin D, Ilijic E, Sun Z, Sampson AR, Mugnaini E, Deutch AY, Sesack SR, Arbuthnott GW, Surmeier DJ (2006) Selective elimination of glutamatergic synapses on striatopallidal neurons in Parkinson disease models. Nat Neurosci 9:251–259PubMedCrossRef
go back to reference De Groot DM (1988) Comparison of methods for the estimation of the thickness of ultrathin tissue sections. J Microsc 151:23–42PubMedCrossRef De Groot DM (1988) Comparison of methods for the estimation of the thickness of ultrathin tissue sections. J Microsc 151:23–42PubMedCrossRef
go back to reference Dempsey D, Jacob P 3rd, Benowitz NL (2000) Nicotine metabolism and elimination kinetics in newborns. Clin Pharmacol Ther 67:458–465PubMedCrossRef Dempsey D, Jacob P 3rd, Benowitz NL (2000) Nicotine metabolism and elimination kinetics in newborns. Clin Pharmacol Ther 67:458–465PubMedCrossRef
go back to reference DiFranza JR, Lew RA (1995) Effect of maternal cigarette smoking on pregnancy complications and sudden infant death syndrome. J Fam Pract 40:385–394PubMed DiFranza JR, Lew RA (1995) Effect of maternal cigarette smoking on pregnancy complications and sudden infant death syndrome. J Fam Pract 40:385–394PubMed
go back to reference Dorph-Petersen KA, Pierri JN, Perel JM, Sun Z, Sampson AR, Lewis DA (2005) The influence of chronic exposure to antipsychotic medications on brain size before and after tissue fixation: a comparison of haloperidol and olanzapine in macaque monkeys. Neuropsychopharmacology 30:1649–1661PubMedCrossRef Dorph-Petersen KA, Pierri JN, Perel JM, Sun Z, Sampson AR, Lewis DA (2005) The influence of chronic exposure to antipsychotic medications on brain size before and after tissue fixation: a comparison of haloperidol and olanzapine in macaque monkeys. Neuropsychopharmacology 30:1649–1661PubMedCrossRef
go back to reference Dwoskin LP, Teng L, Buxton ST, Crooks PA (1999) (S)-(-)-Cotinine, the major brain metabolite of nicotine, stimulates nicotinic receptors to evoke [3H]dopamine release from rat striatal slices in a calcium-dependent manner. J Pharmacol Exp Ther 288:905–911PubMed Dwoskin LP, Teng L, Buxton ST, Crooks PA (1999) (S)-(-)-Cotinine, the major brain metabolite of nicotine, stimulates nicotinic receptors to evoke [3H]dopamine release from rat striatal slices in a calcium-dependent manner. J Pharmacol Exp Ther 288:905–911PubMed
go back to reference El Marroun H, Schmidt MN, Franken IH, Jaddoe VW, Hofman A, van der Lugt A, Verhulst FC, Tiemeier H, White T (2014) Prenatal tobacco exposure and brain morphology: a prospective study in young children. Neuropsychopharmacology 39:792–800PubMedPubMedCentralCrossRef El Marroun H, Schmidt MN, Franken IH, Jaddoe VW, Hofman A, van der Lugt A, Verhulst FC, Tiemeier H, White T (2014) Prenatal tobacco exposure and brain morphology: a prospective study in young children. Neuropsychopharmacology 39:792–800PubMedPubMedCentralCrossRef
go back to reference Ernst M, Moolchan ET, Robinson ML (2001) Behavioral and neural consequences of prenatal exposure to nicotine. J Am Acad Child Adolesc Psychiatry 40:630–641PubMedCrossRef Ernst M, Moolchan ET, Robinson ML (2001) Behavioral and neural consequences of prenatal exposure to nicotine. J Am Acad Child Adolesc Psychiatry 40:630–641PubMedCrossRef
go back to reference Eskenazi B, Castorina R (1999) Association of prenatal maternal or postnatal child environmental tobacco smoke exposure and neurodevelopmental and behavioral problems in children. Environ Health Perspect 107:991–1000PubMedPubMedCentralCrossRef Eskenazi B, Castorina R (1999) Association of prenatal maternal or postnatal child environmental tobacco smoke exposure and neurodevelopmental and behavioral problems in children. Environ Health Perspect 107:991–1000PubMedPubMedCentralCrossRef
go back to reference Francis HW, Rivas A, Lehar M, Saito Y, Mouton PR, Ryugo DK (2006) Efficient quantification of afferent cochlear ultrastructure using design-based stereology. J Neurosci Methods 150:150–158PubMedCrossRef Francis HW, Rivas A, Lehar M, Saito Y, Mouton PR, Ryugo DK (2006) Efficient quantification of afferent cochlear ultrastructure using design-based stereology. J Neurosci Methods 150:150–158PubMedCrossRef
go back to reference Fucile S, Renzi M, Lauro C, Limatola C, Ciotti T, Eusebi F (2004) Nicotinic cholinergic stimulation promotes survival and reduces motility of cultured rat cerebellar granule cells. Neuroscience 127:53–61PubMedCrossRef Fucile S, Renzi M, Lauro C, Limatola C, Ciotti T, Eusebi F (2004) Nicotinic cholinergic stimulation promotes survival and reduces motility of cultured rat cerebellar granule cells. Neuroscience 127:53–61PubMedCrossRef
go back to reference Fung YK (1988) Postnatal behavioural effects of maternal nicotine exposure in rats. J Pharm Pharmacol 40:870–872PubMedCrossRef Fung YK (1988) Postnatal behavioural effects of maternal nicotine exposure in rats. J Pharm Pharmacol 40:870–872PubMedCrossRef
go back to reference Fung YK (1989) Postnatal effects of maternal nicotine exposure on the striatal dopaminergic system in rats. J Pharm Pharmacol 41:576–578PubMedCrossRef Fung YK (1989) Postnatal effects of maternal nicotine exposure on the striatal dopaminergic system in rats. J Pharm Pharmacol 41:576–578PubMedCrossRef
go back to reference Fung YK, Lau YS (1989) Effects of prenatal nicotine exposure on rat striatal dopaminergic and nicotinic systems. Pharmacol Biochem Behav 33:1–6PubMedCrossRef Fung YK, Lau YS (1989) Effects of prenatal nicotine exposure on rat striatal dopaminergic and nicotinic systems. Pharmacol Biochem Behav 33:1–6PubMedCrossRef
go back to reference Geinisman Y, Gundersen HJ, van der Zee E, West MJ (1996) Unbiased stereological estimation of the total number of synapses in a brain region. J Neurocytol 25:805–819PubMedCrossRef Geinisman Y, Gundersen HJ, van der Zee E, West MJ (1996) Unbiased stereological estimation of the total number of synapses in a brain region. J Neurocytol 25:805–819PubMedCrossRef
go back to reference German DC, Manaye KF, Wu D, Hersh LB, Zweig RM (1999) Mesopontine cholinergic and non-cholinergic neurons in schizophrenia. Neuroscience 94:33–38PubMedCrossRef German DC, Manaye KF, Wu D, Hersh LB, Zweig RM (1999) Mesopontine cholinergic and non-cholinergic neurons in schizophrenia. Neuroscience 94:33–38PubMedCrossRef
go back to reference Ghosheh O, Dwoskin LP, Li WK, Crooks PA (1999) Residence times and half-lives of nicotine metabolites in rat brain after acute peripheral administration of [2′-(14)C]nicotine. Drug Metab Dispos 27:1448–1455PubMed Ghosheh O, Dwoskin LP, Li WK, Crooks PA (1999) Residence times and half-lives of nicotine metabolites in rat brain after acute peripheral administration of [2′-(14)C]nicotine. Drug Metab Dispos 27:1448–1455PubMed
go back to reference Godding V, Bonnier C, Fiasse L, Michel M, Longueville E, Lebecque P, Robert A, Galanti L (2004) Does in utero exposure to heavy maternal smoking induce nicotine withdrawal symptoms in neonates? Pediatr Res 55:645–651PubMedCrossRef Godding V, Bonnier C, Fiasse L, Michel M, Longueville E, Lebecque P, Robert A, Galanti L (2004) Does in utero exposure to heavy maternal smoking induce nicotine withdrawal symptoms in neonates? Pediatr Res 55:645–651PubMedCrossRef
go back to reference Greenberg ME, Ziff EB, Greene LA (1986) Stimulation of neuronal acetylcholine receptors induces rapid gene transcription. Science 234:80–83PubMedCrossRef Greenberg ME, Ziff EB, Greene LA (1986) Stimulation of neuronal acetylcholine receptors induces rapid gene transcription. Science 234:80–83PubMedCrossRef
go back to reference Gundersen HJ, Bagger P, Bendtsen TF, Evans SM, Korbo L, Marcussen N, Moller A, Nielsen K, Nyengaard JR, Pakkenberg B et al (1988) The new stereological tools: disector, fractionator, nucleator and point sampled intercepts and their use in pathological research and diagnosis. APMIS 96:857–881PubMedCrossRef Gundersen HJ, Bagger P, Bendtsen TF, Evans SM, Korbo L, Marcussen N, Moller A, Nielsen K, Nyengaard JR, Pakkenberg B et al (1988) The new stereological tools: disector, fractionator, nucleator and point sampled intercepts and their use in pathological research and diagnosis. APMIS 96:857–881PubMedCrossRef
go back to reference Hohmann CF (2003) A morphogenetic role for acetylcholine in mouse cerebral neocortex. Neurosci Biobehav Rev 27:351–363PubMedCrossRef Hohmann CF (2003) A morphogenetic role for acetylcholine in mouse cerebral neocortex. Neurosci Biobehav Rev 27:351–363PubMedCrossRef
go back to reference Ingham CA, Hood SH, Taggart P, Arbuthnott GW (1998) Plasticity of synapses in the rat neostriatum after unilateral lesion of the nigrostriatal dopaminergic pathway. J Neurosci 18:4732–4743PubMed Ingham CA, Hood SH, Taggart P, Arbuthnott GW (1998) Plasticity of synapses in the rat neostriatum after unilateral lesion of the nigrostriatal dopaminergic pathway. J Neurosci 18:4732–4743PubMed
go back to reference Jacob P 3rd, Wilson M, Benowitz NL (1981) Improved gas chromatographic method for the determination of nicotine and cotinine in biologic fluids. J Chromatogr 222:61–70PubMedCrossRef Jacob P 3rd, Wilson M, Benowitz NL (1981) Improved gas chromatographic method for the determination of nicotine and cotinine in biologic fluids. J Chromatogr 222:61–70PubMedCrossRef
go back to reference Jorenby DE (1998) New developments in approaches to smoking cessation. Curr Opin Pulm Med 4:103–106PubMedCrossRef Jorenby DE (1998) New developments in approaches to smoking cessation. Curr Opin Pulm Med 4:103–106PubMedCrossRef
go back to reference Joschko MA, Dreosti IE, Tulsi RS (1991) The teratogenic effects of nicotine in vitro in rats: a light and electron microscope study. Neurotoxicol Teratol 13:307–316PubMedCrossRef Joschko MA, Dreosti IE, Tulsi RS (1991) The teratogenic effects of nicotine in vitro in rats: a light and electron microscope study. Neurotoxicol Teratol 13:307–316PubMedCrossRef
go back to reference Kane VB, Fu Y, Matta SG, Sharp BM (2004) Gestational nicotine exposure attenuates nicotine-stimulated dopamine release in the nucleus accumbens shell of adolescent Lewis rats. J Pharmacol Exp Ther 308:521–528PubMedCrossRef Kane VB, Fu Y, Matta SG, Sharp BM (2004) Gestational nicotine exposure attenuates nicotine-stimulated dopamine release in the nucleus accumbens shell of adolescent Lewis rats. J Pharmacol Exp Ther 308:521–528PubMedCrossRef
go back to reference King BA, Alam S, Promoff G, Arrazola R, Dube SR (2013) Awareness and ever-use of electronic cigarettes among U.S. adults, 2010–2011. Nicotine Tob Res 15:1623–1627PubMedPubMedCentralCrossRef King BA, Alam S, Promoff G, Arrazola R, Dube SR (2013) Awareness and ever-use of electronic cigarettes among U.S. adults, 2010–2011. Nicotine Tob Res 15:1623–1627PubMedPubMedCentralCrossRef
go back to reference Koob GF (1996) Hedonic valence, dopamine and motivation. Mol Psychiatry 1:186–189PubMed Koob GF (1996) Hedonic valence, dopamine and motivation. Mol Psychiatry 1:186–189PubMed
go back to reference Kunzler J, Braun K, Bock J (2013) Early life stress and sex-specific sensitivity of the catecholaminergic systems in prefrontal and limbic regions of Octodon degus. Brain Struct Funct (Epub ahead of print) Kunzler J, Braun K, Bock J (2013) Early life stress and sex-specific sensitivity of the catecholaminergic systems in prefrontal and limbic regions of Octodon degus. Brain Struct Funct (Epub ahead of print)
go back to reference Lambers DS, Clark KE (1996) The maternal and fetal physiologic effects of nicotine. Semin Perinatol 20:115–126PubMedCrossRef Lambers DS, Clark KE (1996) The maternal and fetal physiologic effects of nicotine. Semin Perinatol 20:115–126PubMedCrossRef
go back to reference Leranth C, Roth RH, Elsworth JD, Naftolin F, Horvath TL, Redmond DE Jr (2000) Estrogen is essential for maintaining nigrostriatal dopamine neurons in primates: implications for Parkinson’s disease and memory. J Neurosci 20:8604–8609PubMed Leranth C, Roth RH, Elsworth JD, Naftolin F, Horvath TL, Redmond DE Jr (2000) Estrogen is essential for maintaining nigrostriatal dopamine neurons in primates: implications for Parkinson’s disease and memory. J Neurosci 20:8604–8609PubMed
go back to reference LeSage MG, Gustaf E, Dufek MB, Pentel PR (2006) Effects of maternal intravenous nicotine administration on locomotor behavior in pre-weanling rats. Pharmacol Biochem Behav 85:575–583PubMedPubMedCentralCrossRef LeSage MG, Gustaf E, Dufek MB, Pentel PR (2006) Effects of maternal intravenous nicotine administration on locomotor behavior in pre-weanling rats. Pharmacol Biochem Behav 85:575–583PubMedPubMedCentralCrossRef
go back to reference Lichtensteiger W, Ribary U, Schlumpf M, Odermatt B, Widmer HR (1988) Prenatal adverse effects of nicotine on the developing brain. Prog Brain Res 73:137–157PubMedCrossRef Lichtensteiger W, Ribary U, Schlumpf M, Odermatt B, Widmer HR (1988) Prenatal adverse effects of nicotine on the developing brain. Prog Brain Res 73:137–157PubMedCrossRef
go back to reference Lv J, Mao C, Zhu L, Zhang H, Pengpeng H, Xu F, Liu Y, Zhang L, Xu Z (2008) The effect of prenatal nicotine on expression of nicotine receptor subunits in the fetal brain. Neurotoxicology 29:722–726PubMedPubMedCentralCrossRef Lv J, Mao C, Zhu L, Zhang H, Pengpeng H, Xu F, Liu Y, Zhang L, Xu Z (2008) The effect of prenatal nicotine on expression of nicotine receptor subunits in the fetal brain. Neurotoxicology 29:722–726PubMedPubMedCentralCrossRef
go back to reference Mayhew TM (1992) A review of recent advances in stereology for quantifying neural structure. J Neurocytol 21:313–328PubMedCrossRef Mayhew TM (1992) A review of recent advances in stereology for quantifying neural structure. J Neurocytol 21:313–328PubMedCrossRef
go back to reference McFarland BJ, Seidler FJ, Slotkin TA (1991) Inhibition of DNA synthesis in neonatal rat brain regions caused by acute nicotine administration. Brain Res Dev Brain Res 58:223–229PubMedCrossRef McFarland BJ, Seidler FJ, Slotkin TA (1991) Inhibition of DNA synthesis in neonatal rat brain regions caused by acute nicotine administration. Brain Res Dev Brain Res 58:223–229PubMedCrossRef
go back to reference Messi ML, Renganathan M, Grigorenko E, Delbono O (1997) Activation of alpha7 nicotinic acetylcholine receptor promotes survival of spinal cord motoneurons. FEBS Lett 411:32–38PubMedCrossRef Messi ML, Renganathan M, Grigorenko E, Delbono O (1997) Activation of alpha7 nicotinic acetylcholine receptor promotes survival of spinal cord motoneurons. FEBS Lett 411:32–38PubMedCrossRef
go back to reference Muhammad A, Mychasiuk R, Nakahashi A, Hossain SR, Gibb R, Kolb B (2012) Prenatal nicotine exposure alters neuroanatomical organization of the developing brain. Synapse 66:950–954PubMedCrossRef Muhammad A, Mychasiuk R, Nakahashi A, Hossain SR, Gibb R, Kolb B (2012) Prenatal nicotine exposure alters neuroanatomical organization of the developing brain. Synapse 66:950–954PubMedCrossRef
go back to reference Muneoka K, Ogawa T, Kamei K, Muraoka S, Tomiyoshi R, Mimura Y, Kato H, Suzuki MR, Takigawa M (1997) Prenatal nicotine exposure affects the development of the central serotonergic system as well as the dopaminergic system in rat offspring: involvement of route of drug administrations. Brain Res Dev Brain Res 102:117–126PubMedCrossRef Muneoka K, Ogawa T, Kamei K, Muraoka S, Tomiyoshi R, Mimura Y, Kato H, Suzuki MR, Takigawa M (1997) Prenatal nicotine exposure affects the development of the central serotonergic system as well as the dopaminergic system in rat offspring: involvement of route of drug administrations. Brain Res Dev Brain Res 102:117–126PubMedCrossRef
go back to reference Muneoka K, Nakatsu T, Fuji J, Ogawa T, Takigawa M (1999) Prenatal administration of nicotine results in dopaminergic alterations in the neocortex. Neurotoxicol Teratol 21:603–609PubMedCrossRef Muneoka K, Nakatsu T, Fuji J, Ogawa T, Takigawa M (1999) Prenatal administration of nicotine results in dopaminergic alterations in the neocortex. Neurotoxicol Teratol 21:603–609PubMedCrossRef
go back to reference Mychasiuk R, Muhammad A, Carroll C, Kolb B (2013a) Does prenatal nicotine exposure alter the brain’s response to nicotine in adolescence? A neuroanatomical analysis. Eur J Neurosci 38:2491–2503PubMedCrossRef Mychasiuk R, Muhammad A, Carroll C, Kolb B (2013a) Does prenatal nicotine exposure alter the brain’s response to nicotine in adolescence? A neuroanatomical analysis. Eur J Neurosci 38:2491–2503PubMedCrossRef
go back to reference Mychasiuk R, Muhammad A, Gibb R, Kolb B (2013b) Long-term alterations to dendritic morphology and spine density associated with prenatal exposure to nicotine. Brain Res 1499:53–60PubMedCrossRef Mychasiuk R, Muhammad A, Gibb R, Kolb B (2013b) Long-term alterations to dendritic morphology and spine density associated with prenatal exposure to nicotine. Brain Res 1499:53–60PubMedCrossRef
go back to reference Naeff B, Schlumpf M, Lichtensteiger W (1992) Pre- and postnatal development of high-affinity [3H]nicotine binding sites in rat brain regions: an autoradiographic study. Dev Brain Res 68:163–174CrossRef Naeff B, Schlumpf M, Lichtensteiger W (1992) Pre- and postnatal development of high-affinity [3H]nicotine binding sites in rat brain regions: an autoradiographic study. Dev Brain Res 68:163–174CrossRef
go back to reference Nair-Roberts RG, Chatelain-Badie SD, Benson E, White-Cooper H, Bolam JP, Ungless MA (2008) Stereological estimates of dopaminergic, GABAergic and glutamatergic neurons in the ventral tegmental area, substantia nigra and retrorubral field in the rat. Neuroscience 152:1024–1031PubMedPubMedCentralCrossRef Nair-Roberts RG, Chatelain-Badie SD, Benson E, White-Cooper H, Bolam JP, Ungless MA (2008) Stereological estimates of dopaminergic, GABAergic and glutamatergic neurons in the ventral tegmental area, substantia nigra and retrorubral field in the rat. Neuroscience 152:1024–1031PubMedPubMedCentralCrossRef
go back to reference Navarro HA, Seidler FJ, Whitmore WL, Slotkin TA (1988) Prenatal exposure to nicotine via maternal infusions: effects on development of catecholamine systems. J Pharmacol Exp Ther 244:940–944PubMed Navarro HA, Seidler FJ, Whitmore WL, Slotkin TA (1988) Prenatal exposure to nicotine via maternal infusions: effects on development of catecholamine systems. J Pharmacol Exp Ther 244:940–944PubMed
go back to reference Newman MB, Shytle RD, Sanberg PR (1999) Locomotor behavioral effects of prenatal and postnatal nicotine exposure in rat offspring. Behav Pharmacol 10:699–706PubMedCrossRef Newman MB, Shytle RD, Sanberg PR (1999) Locomotor behavioral effects of prenatal and postnatal nicotine exposure in rat offspring. Behav Pharmacol 10:699–706PubMedCrossRef
go back to reference Nguyen L, Rigo JM, Rocher V, Belachew S, Malgrange B, Rogister B, Leprince P, Moonen G (2001) Neurotransmitters as early signals for central nervous system development. Cell Tissue Res 305:187–202PubMedCrossRef Nguyen L, Rigo JM, Rocher V, Belachew S, Malgrange B, Rogister B, Leprince P, Moonen G (2001) Neurotransmitters as early signals for central nervous system development. Cell Tissue Res 305:187–202PubMedCrossRef
go back to reference Niaura R, Bock B, Lloyd EE, Brown R, Lipsitt LP, Buka S (2001) Maternal transmission of nicotine dependence: psychiatric, neurocognitive and prenatal factors. Am J Addict 10:16–29PubMedCrossRef Niaura R, Bock B, Lloyd EE, Brown R, Lipsitt LP, Buka S (2001) Maternal transmission of nicotine dependence: psychiatric, neurocognitive and prenatal factors. Am J Addict 10:16–29PubMedCrossRef
go back to reference Omelchenko N, Sesack SR (2005) Laterodorsal tegmental projections to identified cell populations in the rat ventral tegmental area. J Comp Neurol 483:217–235PubMedCrossRef Omelchenko N, Sesack SR (2005) Laterodorsal tegmental projections to identified cell populations in the rat ventral tegmental area. J Comp Neurol 483:217–235PubMedCrossRef
go back to reference Omelchenko N, Sesack SR (2006) Cholinergic axons in the rat ventral tegmental area synapse preferentially onto mesoaccumbens dopamine neurons. J Comp Neurol 494:863–875PubMedPubMedCentralCrossRef Omelchenko N, Sesack SR (2006) Cholinergic axons in the rat ventral tegmental area synapse preferentially onto mesoaccumbens dopamine neurons. J Comp Neurol 494:863–875PubMedPubMedCentralCrossRef
go back to reference Omelchenko N, Sesack SR (2007) Glutamate synaptic inputs to ventral tegmental area neurons in the rat derive primarily from subcortical sources. Neuroscience 146:1259–1274PubMedPubMedCentralCrossRef Omelchenko N, Sesack SR (2007) Glutamate synaptic inputs to ventral tegmental area neurons in the rat derive primarily from subcortical sources. Neuroscience 146:1259–1274PubMedPubMedCentralCrossRef
go back to reference Omelchenko N, Bell R, Sesack SR (2009) Lateral habenula projections to dopamine and GABA neurons in the rat ventral tegmental area. Eur J Neurosci 30:1239–1250PubMedPubMedCentralCrossRef Omelchenko N, Bell R, Sesack SR (2009) Lateral habenula projections to dopamine and GABA neurons in the rat ventral tegmental area. Eur J Neurosci 30:1239–1250PubMedPubMedCentralCrossRef
go back to reference Onal A, Uysal A, Ulker S, Delen Y, Yurtseven ME, Evinc A (2004) Alterations of brain tissue in fetal rats exposed to nicotine in utero: possible involvement of nitric oxide and catecholamines. Neurotoxicol Teratol 26:103–112PubMedCrossRef Onal A, Uysal A, Ulker S, Delen Y, Yurtseven ME, Evinc A (2004) Alterations of brain tissue in fetal rats exposed to nicotine in utero: possible involvement of nitric oxide and catecholamines. Neurotoxicol Teratol 26:103–112PubMedCrossRef
go back to reference Pauly JR, Sparks JA, Hauser KF, Pauly TH (2004) In utero nicotine exposure causes persistent, gender-dependant changes in locomotor activity and sensitivity to nicotine in C57Bl/6 mice. Int J Dev Neurosci 22:329–337PubMedCrossRef Pauly JR, Sparks JA, Hauser KF, Pauly TH (2004) In utero nicotine exposure causes persistent, gender-dependant changes in locomotor activity and sensitivity to nicotine in C57Bl/6 mice. Int J Dev Neurosci 22:329–337PubMedCrossRef
go back to reference Paxinos G, Watson C (1998) The rat brain in stereotaxic coordinates. Academic Press, New York Paxinos G, Watson C (1998) The rat brain in stereotaxic coordinates. Academic Press, New York
go back to reference Peters A, Palay SL, Webster HF (1991) The fine structure of the nervous system. Neurons and their supporting cells. Oxford University Press, New York Peters A, Palay SL, Webster HF (1991) The fine structure of the nervous system. Neurons and their supporting cells. Oxford University Press, New York
go back to reference Phillipson OT (1979) Afferent projections to the ventral tegmental area of Tsai and interfascicular nucleus: a horseradish peroxidase study in the rat. J Comp Neurol 187:117–144PubMedCrossRef Phillipson OT (1979) Afferent projections to the ventral tegmental area of Tsai and interfascicular nucleus: a horseradish peroxidase study in the rat. J Comp Neurol 187:117–144PubMedCrossRef
go back to reference Pichini S, Basagana XB, Pacifici R, Garcia O, Puig C, Vall O, Harris J, Zuccaro P, Segura J, Sunyer J (2000) Cord serum cotinine as a biomarker of fetal exposure to cigarette smoke at the end of pregnancy. Environ Health Perspect 108:1079–1083PubMedPubMedCentralCrossRef Pichini S, Basagana XB, Pacifici R, Garcia O, Puig C, Vall O, Harris J, Zuccaro P, Segura J, Sunyer J (2000) Cord serum cotinine as a biomarker of fetal exposure to cigarette smoke at the end of pregnancy. Environ Health Perspect 108:1079–1083PubMedPubMedCentralCrossRef
go back to reference Qiao D, Seidler FJ, Violin JD, Slotkin TA (2003) Nicotine is a developmental neurotoxicant and neuroprotectant: stage-selective inhibition of DNA synthesis coincident with shielding from effects of chlorpyrifos. Brain Res Dev Brain Res 147:183–190PubMedCrossRef Qiao D, Seidler FJ, Violin JD, Slotkin TA (2003) Nicotine is a developmental neurotoxicant and neuroprotectant: stage-selective inhibition of DNA synthesis coincident with shielding from effects of chlorpyrifos. Brain Res Dev Brain Res 147:183–190PubMedCrossRef
go back to reference Redgrave P, Prescott TJ, Gurney K (1999) Is the short-latency dopamine response too short to signal reward error? Trends Neurosci 22:146–151PubMedCrossRef Redgrave P, Prescott TJ, Gurney K (1999) Is the short-latency dopamine response too short to signal reward error? Trends Neurosci 22:146–151PubMedCrossRef
go back to reference Ribary U, Lichtensteiger W (1989) Effects of acute and chronic prenatal nicotine treatment on central catecholamine systems of male and female rat fetuses and offspring. J Pharmacol Exp Ther 248:786–792PubMed Ribary U, Lichtensteiger W (1989) Effects of acute and chronic prenatal nicotine treatment on central catecholamine systems of male and female rat fetuses and offspring. J Pharmacol Exp Ther 248:786–792PubMed
go back to reference Richardson SA, Tizabi Y (1994) Hyperactivity in the offspring of nicotine-treated rats: role of the mesolimbic and nigrostriatal dopaminergic pathways. Pharmacol Biochem Behav 47:331–337PubMedCrossRef Richardson SA, Tizabi Y (1994) Hyperactivity in the offspring of nicotine-treated rats: role of the mesolimbic and nigrostriatal dopaminergic pathways. Pharmacol Biochem Behav 47:331–337PubMedCrossRef
go back to reference Romero RD, Chen WJ (2004) Gender-related response in open-field activity following developmental nicotine exposure in rats. Pharmacol Biochem Behav 78:675–681PubMedCrossRef Romero RD, Chen WJ (2004) Gender-related response in open-field activity following developmental nicotine exposure in rats. Pharmacol Biochem Behav 78:675–681PubMedCrossRef
go back to reference Romijn HJ, Hofman MA, Gramsbergen A (1991) At what age is the developing cerebral cortex of the rat comparable to that of the full-term newborn human baby? Early Hum Dev 26:61–67PubMedCrossRef Romijn HJ, Hofman MA, Gramsbergen A (1991) At what age is the developing cerebral cortex of the rat comparable to that of the full-term newborn human baby? Early Hum Dev 26:61–67PubMedCrossRef
go back to reference Roy TS, Sabherwal U (1994) Effects of prenatal nicotine exposure on the morphogenesis of somatosensory cortex. Neurotoxicol Teratol 16:411–421PubMedCrossRef Roy TS, Sabherwal U (1994) Effects of prenatal nicotine exposure on the morphogenesis of somatosensory cortex. Neurotoxicol Teratol 16:411–421PubMedCrossRef
go back to reference Roy TS, Sabherwal U (1998) Effects of gestational nicotine exposure on hippocampal morphology. Neurotoxicol Teratol 20:465–473PubMedCrossRef Roy TS, Sabherwal U (1998) Effects of gestational nicotine exposure on hippocampal morphology. Neurotoxicol Teratol 20:465–473PubMedCrossRef
go back to reference Roy TS, Seidler FJ, Slotkin TA (2002) Prenatal nicotine exposure evokes alterations of cell structure in hippocampus and somatosensory cortex. J Pharmacol Exp Ther 300:124–133PubMedCrossRef Roy TS, Seidler FJ, Slotkin TA (2002) Prenatal nicotine exposure evokes alterations of cell structure in hippocampus and somatosensory cortex. J Pharmacol Exp Ther 300:124–133PubMedCrossRef
go back to reference Saal D, Dong Y, Bonci A, Malenka RC (2003) Drugs of abuse and stress trigger a common synaptic adaptation in dopamine neurons. Neuron 37:577–582PubMedCrossRef Saal D, Dong Y, Bonci A, Malenka RC (2003) Drugs of abuse and stress trigger a common synaptic adaptation in dopamine neurons. Neuron 37:577–582PubMedCrossRef
go back to reference Santiago SE, Huffman KJ (2012) Postnatal effects of prenatal nicotine exposure on body weight, brain size and cortical connectivity in mice. Neurosci Res 73:282–291PubMedCrossRef Santiago SE, Huffman KJ (2012) Postnatal effects of prenatal nicotine exposure on body weight, brain size and cortical connectivity in mice. Neurosci Res 73:282–291PubMedCrossRef
go back to reference Scalera A, Koren G (1998) Rationale for treating pregnant smokers with nicotine patches. Can Fam Phys 44:1601–1603 Scalera A, Koren G (1998) Rationale for treating pregnant smokers with nicotine patches. Can Fam Phys 44:1601–1603
go back to reference Schneider AS, Atluri P, Shen Q, Barnes W, Mah SJ, Stadfelt D, Goderie SK, Temple S, Fleck MW (2002) Functional nicotinic acetylcholine receptor expression on stem and progenitor cells of the early embryonic nervous system. Ann N Y Acad Sci 971:135–138PubMedCrossRef Schneider AS, Atluri P, Shen Q, Barnes W, Mah SJ, Stadfelt D, Goderie SK, Temple S, Fleck MW (2002) Functional nicotinic acetylcholine receptor expression on stem and progenitor cells of the early embryonic nervous system. Ann N Y Acad Sci 971:135–138PubMedCrossRef
go back to reference Schneider T, Ilott N, Brolese G, Bizarro L, Asherson PJ, Stolerman IP (2011) Prenatal exposure to nicotine impairs performance of the 5-choice serial reaction time task in adult rats. Neuropsychopharmacology 36:1114–1125PubMedPubMedCentralCrossRef Schneider T, Ilott N, Brolese G, Bizarro L, Asherson PJ, Stolerman IP (2011) Prenatal exposure to nicotine impairs performance of the 5-choice serial reaction time task in adult rats. Neuropsychopharmacology 36:1114–1125PubMedPubMedCentralCrossRef
go back to reference Seidler FJ, Albright ES, Lappi SE, Slotkin TA (1994) In search of a mechanism for receptor-mediated neurobehavioral teratogenesis by nicotine: catecholamine release by nicotine in immature rat brain regions. Dev Brain Res 82:1–8CrossRef Seidler FJ, Albright ES, Lappi SE, Slotkin TA (1994) In search of a mechanism for receptor-mediated neurobehavioral teratogenesis by nicotine: catecholamine release by nicotine in immature rat brain regions. Dev Brain Res 82:1–8CrossRef
go back to reference Sesack SR, Snyder CL, Lewis DA (1995) Axon terminals immunolabeled for dopamine or tyrosine hydroxylase synapse on GABA-immunoreactive dendrites in rat and monkey cortex. J Comp Neurol 363:264–280PubMedCrossRef Sesack SR, Snyder CL, Lewis DA (1995) Axon terminals immunolabeled for dopamine or tyrosine hydroxylase synapse on GABA-immunoreactive dendrites in rat and monkey cortex. J Comp Neurol 363:264–280PubMedCrossRef
go back to reference Shenassa ED, McCaffery JM, Swan GE, Khroyan TV, Shakib S, Lerman C, Lyons M, Mouttapa M, Niaura RS, Buka SL, Leslie F, Santangelo SL (2003) Intergenerational transmission of tobacco use and dependence: a transdisciplinary perspective. Nicotine Tob Res 5(Suppl 1):S55–69PubMedCrossRef Shenassa ED, McCaffery JM, Swan GE, Khroyan TV, Shakib S, Lerman C, Lyons M, Mouttapa M, Niaura RS, Buka SL, Leslie F, Santangelo SL (2003) Intergenerational transmission of tobacco use and dependence: a transdisciplinary perspective. Nicotine Tob Res 5(Suppl 1):S55–69PubMedCrossRef
go back to reference Slotkin TA (1998) Fetal nicotine or cocaine exposure: which one is worse? J Pharmacol Exp Ther 285:931–945PubMed Slotkin TA (1998) Fetal nicotine or cocaine exposure: which one is worse? J Pharmacol Exp Ther 285:931–945PubMed
go back to reference Slotkin TA, Cho H, Whitmore WL (1987) Effects of prenatal nicotine exposure on neuronal development: selective actions on central and peripheral catecholaminergic pathways. Brain Res Bull 18:601–611PubMedCrossRef Slotkin TA, Cho H, Whitmore WL (1987) Effects of prenatal nicotine exposure on neuronal development: selective actions on central and peripheral catecholaminergic pathways. Brain Res Bull 18:601–611PubMedCrossRef
go back to reference Slotkin TA, McCook EC, Seidler FJ (1997) Cryptic brain cell injury caused by fetal nicotine exposure is associated with persistent elevations of c-fos protooncogene expression. Brain Res 750:180–188PubMedCrossRef Slotkin TA, McCook EC, Seidler FJ (1997) Cryptic brain cell injury caused by fetal nicotine exposure is associated with persistent elevations of c-fos protooncogene expression. Brain Res 750:180–188PubMedCrossRef
go back to reference Swanson LW (1982) The projections of the ventral tegmental area and adjacent regions: a combined fluorescent retrograde tracer and immunofluorescence study in the rat. Brain Res Bull 9:321–353PubMedCrossRef Swanson LW (1982) The projections of the ventral tegmental area and adjacent regions: a combined fluorescent retrograde tracer and immunofluorescence study in the rat. Brain Res Bull 9:321–353PubMedCrossRef
go back to reference Thomas JD, Garrison ME, Slawecki CJ, Ehlers CL, Riley EP (2000) Nicotine exposure during the neonatal brain growth spurt produces hyperactivity in preweanling rats. Neurotoxicol Teratol 22:695–701PubMedCrossRef Thomas JD, Garrison ME, Slawecki CJ, Ehlers CL, Riley EP (2000) Nicotine exposure during the neonatal brain growth spurt produces hyperactivity in preweanling rats. Neurotoxicol Teratol 22:695–701PubMedCrossRef
go back to reference Tizabi Y, Popke EJ, Rahman MA, Nespor SM, Grunberg NE (1997) Hyperactivity induced by prenatal nicotine exposure is associated with an increase in cortical nicotinic receptors. Pharmacol Biochem Behav 58:141–146PubMedCrossRef Tizabi Y, Popke EJ, Rahman MA, Nespor SM, Grunberg NE (1997) Hyperactivity induced by prenatal nicotine exposure is associated with an increase in cortical nicotinic receptors. Pharmacol Biochem Behav 58:141–146PubMedCrossRef
go back to reference Tizabi Y, Russell LT, Nespor SM, Perry DC, Grunberg NE (2000) Prenatal nicotine exposure: effects on locomotor activity and central [125I]alpha-BT binding in rats. Pharmacol Biochem Behav 66:495–500PubMedCrossRef Tizabi Y, Russell LT, Nespor SM, Perry DC, Grunberg NE (2000) Prenatal nicotine exposure: effects on locomotor activity and central [125I]alpha-BT binding in rats. Pharmacol Biochem Behav 66:495–500PubMedCrossRef
go back to reference Trauth JA, Seidler FJ, McCook EC, Slotkin TA (1999) Persistent c-fos induction by nicotine in developing rat brain regions: interaction with hypoxia. Pediatr Res 45:38–45PubMedCrossRef Trauth JA, Seidler FJ, McCook EC, Slotkin TA (1999) Persistent c-fos induction by nicotine in developing rat brain regions: interaction with hypoxia. Pediatr Res 45:38–45PubMedCrossRef
go back to reference Trauth JA, Seidler FJ, Slotkin TA (2000) An animal model of adolescent nicotine exposure: effects on gene expression and macromolecular constituents in rat brain regions. Brain Res 867:29–39PubMedCrossRef Trauth JA, Seidler FJ, Slotkin TA (2000) An animal model of adolescent nicotine exposure: effects on gene expression and macromolecular constituents in rat brain regions. Brain Res 867:29–39PubMedCrossRef
go back to reference Tribollet E, Bertrand D, Marguerat A, Raggenbass M (2004) Comparative distribution of nicotinic receptor subtypes during development, adulthood and aging: an autoradiographic study in the rat brain. Neuroscience 124:405–420PubMedCrossRef Tribollet E, Bertrand D, Marguerat A, Raggenbass M (2004) Comparative distribution of nicotinic receptor subtypes during development, adulthood and aging: an autoradiographic study in the rat brain. Neuroscience 124:405–420PubMedCrossRef
go back to reference Vaglenova J, Birru S, Pandiella NM, Breese CR (2004) An assessment of the long-term developmental and behavioral teratogenicity of prenatal nicotine exposure. Behav Brain Res 150:159–170PubMedCrossRef Vaglenova J, Birru S, Pandiella NM, Breese CR (2004) An assessment of the long-term developmental and behavioral teratogenicity of prenatal nicotine exposure. Behav Brain Res 150:159–170PubMedCrossRef
go back to reference Woolley CS, McEwen BS (1992) Estradiol mediates fluctuation in hippocampal synapse density during the estrous cycle in the adult rat. J Neurosci 12:2549–2554PubMed Woolley CS, McEwen BS (1992) Estradiol mediates fluctuation in hippocampal synapse density during the estrous cycle in the adult rat. J Neurosci 12:2549–2554PubMed
go back to reference Zhang X, Liu C, Miao H, Gong ZH, Nordberg A (1998) Postnatal changes of nicotinic acetylcholine receptor alpha 2, alpha 3, alpha 4, alpha 7 and beta 2 subunits genes expression in rat brain. Int J Dev Neurosci 16:507–518PubMedCrossRef Zhang X, Liu C, Miao H, Gong ZH, Nordberg A (1998) Postnatal changes of nicotinic acetylcholine receptor alpha 2, alpha 3, alpha 4, alpha 7 and beta 2 subunits genes expression in rat brain. Int J Dev Neurosci 16:507–518PubMedCrossRef
go back to reference Zoli M, Le Novere N, Hill JA Jr, Changeux JP (1995) Developmental regulation of nicotinic ACh receptor subunit mRNAs in the rat central and peripheral nervous systems. J Neurosci 15:1912–1939PubMed Zoli M, Le Novere N, Hill JA Jr, Changeux JP (1995) Developmental regulation of nicotinic ACh receptor subunit mRNAs in the rat central and peripheral nervous systems. J Neurosci 15:1912–1939PubMed
Metadata
Title
Impact of prenatal nicotine on the structure of midbrain dopamine regions in the rat
Authors
Natalia Omelchenko
Priya Roy
Judith Joyce Balcita-Pedicino
Samuel Poloyac
Susan R. Sesack
Publication date
01-05-2016
Publisher
Springer Berlin Heidelberg
Published in
Brain Structure and Function / Issue 4/2016
Print ISSN: 1863-2653
Electronic ISSN: 1863-2661
DOI
https://doi.org/10.1007/s00429-015-1014-y

Other articles of this Issue 4/2016

Brain Structure and Function 4/2016 Go to the issue