Abstract
Addiction is a complex disorder because many factors contribute to the development and maintenance of addiction. One factor is learning. For example, drug–context associations that develop during drug use could facilitate drug craving upon re-exposure to contexts previously associated with drugs. Additionally, deficits in cognitive processes associated with withdrawal could precipitate relapse in attempts to ameliorate those deficits. Because addiction and learning involve common neural areas and cell signaling cascades, addiction-related changes in processes underlying plasticity may contribute to addiction. This article examines similarities between addiction and learning at the behavioral, neural, and cellular levels, with emphasis on the neural substrates underlying the effects of acute nicotine, chronic nicotine, and withdrawal from chronic nicotine on hippocampus-dependent contextual learning.
Index Entries: Learning, addiction, acetylcholine, nicotine, hippocampus, contextual fear conditioning, CREB, MAPK, withdrawal, plasticity
Introduction
Despite overwhelming evidence for the adverse health effects of smoking, 68.8 million Americans use tobacco products and 400,000 tobacco-related deaths occur in the United States each year (1). Animal models have shown that nicotine has strong reinforcing properties, that abstinence from nicotine after chronic nicotine treatment produces withdrawal symptoms, and that exposure to environments where drug use occurs result in reinstatement of nicotine-seeking behavior (for review, see refs. 2–4). Nicotine withdrawal studies with rodents have identified multiple withdrawal-related changes in somatic and affective responses. In both mice and rats, nicotine withdrawal resulted in the expression of somatic withdrawal behaviors such as head shakes and paw tremors (5,6). Additionally, nicotine withdrawal was shown to be anxiogenic in mice (6,7) and was associated with a marked decrease in brain reward function in rats (8). Nicotine withdrawal also disrupted cognition-related functioning in rodents, as measured by the ability to sensory-gate stimuli (9). However, this effect may be dose-dependent (7).
In humans, nicotine withdrawal is associated with anger, anxiety, difficulty concentrating, increased appetite, and agitation (10). Additionally, smoking cessation is associated with disruption of sensory gating (11–13) and cognitive function (14–16). The reduction of negative symptoms associated with nicotine withdrawal may motivate continued tobacco use and relapse (17). However, the expression and severity of withdrawal symptoms may depend on multiple factors, including age, genetics, environment, and mental health (17–21). Therefore, there may not be a universal symptom of nicotine withdrawal. Rather, a range of symptoms, including disrupted cognition, may characterize nicotine withdrawal. Increasing efforts have focused not only on understanding the effects of withdrawal on cognitive processes (e.g., learning) but also on understanding the role of learning in addiction. This article examines the role of learning and memory in addiction, with emphasis on the neural substrates underlying the effects of acute nicotine, chronic nicotine, and withdrawal from chronic nicotine on hippocampus-dependent contextual learning.
Neural and Cellular Correlates of Learning, Memory, and Addiction
In addition to the reinforcing properties of nicotine and the somatic withdrawal symptoms that help to maintain nicotine addiction, changes in the neural substrates of learning and memory may also contribute to the development and maintenance of addiction. Tremendous overlap exists between the neural and cellular substrates of learning and the neural and cellular substrates of addiction. The ability of drugs of abuse to interact with and alter the neural substrates of learning may contribute to the strong addictive properties of these drugs. Furthermore, drugs of abuse may have an especially strong impact on the declarative memory system. The declarative memory system allows for recollection of facts and events (22); these types of memories comprise the personal history of individuals. Neural areas involved in declarative memory include the medial prefrontal cortex, hippocampus, parahippocampal regions, amygdala, and nucleus accumbens (23–27). These areas are also involved in addiction. For example, addiction is associated with alterations in cortical function that may lead to disrupted decision processes and compulsive drug use (28,29). Furthermore, Bechara (30) proposed that the amygdala may become hypersensitive to reward during addiction, and this overactivation of the amygdala may lead to altered regulation of ventromedial prefrontal cortical activity involved in decision making. Additionally, numerous studies have shown involvement of the amygdala in the formation of drug–cue associations (for review, see ref. 31) and involvement of the nucleus accumbens, which is part of the reward circuitry (32), in drug-seeking behavior (33–35).
Finally, the hippocampus may process contextual drug associations that contribute to context-evoked craving and drug-seeking behavior. Inactivation of the hippocampus with tetrodotoxin prevented context-stimulated reinstatement of cocaine-seeking behavior (36), and θ-wave stimulation of the hippocampus produced cocaine-seeking behavior (37). Although this is not a complete list of brain areas involved in addiction, multiple groups have proposed that these areas form an interconnected system involved in drug addiction (31,38–41). Therefore, because the declarative memory system is critically involved in processing and storing the memories that provide individuals with a personal history, the ability of drugs of abuse to co-opt the declarative memory system may partially explain the strong addictive nature of these drugs.
Just as learning and addiction share similar neural substrates, learning and addiction also share similar cellular and molecular substrates. Tremendous headway has been made in identifying the cell signaling cascades involved in learning and synaptic plasticity. Briefly, numerous studies have demonstrated that protein kinase A (PKA), mitogen-activated protein kinase/extracellular-regulated kinase (MAPK/ERK), calcium/calmodulin protein kinase II (CaMKII), and the gene transcription factor cyclic adenosine monophosphate (cAMP)-response-element-binding protein (CREB) are critically involved in learning and synaptic plasticity (42–47). These same substrates are involved in addiction. Infusion of a PKA inhibitor into the nucleus accumbens decreased cocaine self-administration, and infusion of a PKA activator increased self-administration of cocaine in rats (48,49). In Drosophila, sensitization after repeated nicotine exposure was associated with increased levels of cAMP, implicating the cAMP/PKA pathway in this behavioral effect (50). Furthermore, infusion of either a CaMKII inhibitor into the nucleus accumbens or an ERK inhibitor into the ventral tegmental area disrupted the development of behavioral sensitization to cocaine (51,52), and inhibition of ERK phosphorylation in the central amygdala decreased cocaine-seeking behavior (53). Changes in ERK activation may also contribute to alcoholism; withdrawal from ethanol was associated with increased ERK activation in multiple brain regions, including the amygdala and hippocampus (54).
Changes in kinase levels can lead to changes in gene expression. The gene transcription factor CREB is activated by multiple kinases, including PKA and MAPK (for review, see refs. 55 and 56). Therefore, the involvement of PKA and MAPK in addiction suggests a role for CREB as well. Evidence for the involvement of CREB in addiction comes from studies demonstrating that withdrawal from nicotine is associated with altered levels of phosphorylated and total CREB in the nucleus accumbens (57), inhibition of CREB in the nucleus accumbens increased the rewarding properties of cocaine (58), and chronic morphine administration decreased levels of CREB in the nucleus accumbens (59). Therefore, learning and addiction activate similar cell signaling cascades and neural areas, and the ability of drugs of abuse to alter cell-signaling cascades involved in synaptic plasticity may lead to long-lasting behavioral changes. It is important to determine how activation of cell signaling cascades changes as drug administration changes from acute to chronic to withdrawal as well as to determine if activation and expression patterns vary across brain regions.
Learning and Memory and Nicotine Addiction
Similarly to other drugs of abuse, learning contributes to development and maintenance of nicotine addiction. Multiple studies have demonstrated that conditioned place preference can be established for a context associated with nicotine administration (60–65). Whereas factors such as genetics and age may influence the development of conditioned place preference to nicotine, these studies clearly show that associations between the effects of nicotine and an environment can be conditioned. The ability to form strong but maladaptive associations between drug use and contextual stimuli may contribute to addiction and relapse by triggering context-specific drug craving and drug seeking. Specifically, researchers have proposed that nicotine reinforcement involves enhancement of associations with non-nicotine stimuli that eventually become reinforcers (66). These non-nicotine stimuli could contribute to continued nicotine use. Smoking can be maintained by environmental stimuli (67,68), and acquisition of nicotine self-administration in animal models is enhanced by pairing nicotine delivery with non-nicotine stimuli (69–72). Additionally, during extinction of nicotine self-administration, the presence of a non-nicotine stimulus previously paired with nicotine sustains pressing of a lever previously associated with nicotine (67,72). These data suggest that these learned associations may contribute to resistance to extinction of nicotine self-administration. Furthermore, these studies together provide strong evidence for the role of learning in nicotine addiction.
Research demonstrating that environmental stimuli impact addiction (36) and the potential involvement of declarative memory processes in addiction (73) has led our lab to examine the effects of nicotine on contextual fear conditioning, a form of hippocampus-dependent learning (74,75) that may model declarative memory processes. Nicotine has multiple effects on learning that may contribute to the development and maintenance of addiction to nicotine. Acute nicotine, which may model the initial effects of smoking, enhances learning (76–81). This positive effect of nicotine could facilitate continued drug use. Additionally, cognitive enhancing properties of nicotine could facilitate the formation of maladaptive drug–context associations that can lead to context-evoked cravings. Furthermore, if tolerance to the cognitive enhancing effects of nicotine develops, then greater amounts of nicotine may be consumed to compensate. This increased consumption could increase the addictive liability of nicotine. Finally, because withdrawal is associated with deficits in cognition, relapse may occur to ameliorate these deficits. Our work has focused on understanding the effects of acute nicotine treatment, chronic nicotine treatment, and withdrawal from chronic nicotine treatment on contextual conditioning and on the underlying nicotinic acetylcholinergic receptor (nAChR) subtypes and cell signaling cascades mediating the effects of nicotine on contextual fear conditioning. Because nAChR subtypes have different functional properties, a brief review of nAChRs follows to preface the discussion of behavioral effects of nicotine and the underlying neural substrates that may be involved.
Nicotinic Acetylcholinergic Receptor
nAChRs are a class of ligand-gated ion channels assembled from five subunits. Seventeen identified subunits that are differentially expressed in the central nervous system and peripheral nervous system have been identified (82–85). Whereas the α4β2 and α7 nAChRs are similar because they are the predominant nAChR subtypes in the central nervous system, they differ in their functional properties (86–88). α4β2 nAChR subtypes show high affinity for nicotine, desensitize slowly, and show long-lasting inhibition by mecamylamine, a broad spectrum nAChR antagonist (85,89,90). Conversely, α7 nAChR subtypes show lower affinity for nicotine and a high affinity for α-bungarotoxin, desensitize rapidly, and show shortlasting inhibition by mecamylamine (85, 89,91–94). Because nAChRs have different functional properties, nAChR subtypes may differentially contribute to the effects of nicotine on learning and addiction.
Both the α7 and α4β2 nAChR subtypes have properties that could contribute to cellular changes associated with learning and addiction. Both subtypes are located in the hippocampus, and α4β2 and α7 nAChRs are expressed pre and postsynaptically, suggesting that these receptor subtypes could modulate both pre- and postsynaptic processes involved in synaptic plasticity. Furthmore, α4β2 and α7 nAChRs are calcium-permeable, which could enhance activation of second messengers involved in synaptic plasticity (86–88,94–103). Some studies have suggested that the α7 and α4β2 nAChR subtypes may mediate different behavioral processes (104,105). For example, activation of non-α7 nAChRs enhanced long-term potentiation, and inhibition of α7 nAChR subtypes enhanced long-term potentiation (106). These data suggest that nicotine binding at these nAChR subtypes may differentially affect learning processes. Understanding the role of nAChR subtypes in the effects of acute, chronic, and withdrawal from chronic nicotine aids in understanding the behavioral effects of nicotine from receptor activation to molecular changes.
Acute Nicotine
We have used fear conditioning to examine the behavioral and neural effects of acute nicotine administration on contextual and noncontextual learning. In fear conditioning, animals form an association between a discrete auditory conditioned stimulus (CS) and a foot-shock unconditioned stimulus (US; i.e., cued fear conditioning) and between the training context and the US (i.e., contextual fear conditioning). Learning to associate the context with the US is hippocampus- and amygdala-dependent (74,75). On the other hand, learning to associate the auditory CS with the foot-shock US involves many of the same brain regions as contextual fear conditioning but not the hippocampus (75). Therefore, fear conditioning allows for the assessment of both hippocampus-dependent and hippocampus-independent learning in the same animal after a single training session.
A large body of work has examined both the neurocircuitry (reviewed in ref. 107) and the associated mechanisms of plasticity (e.g., ref. 55) that supports contextual fear conditioning, making contextual fear conditioning an excellent behavioral paradigm for examining the effects of nicotine on hippocampus-dependent learning. We have shown that acute nicotine enhances contextual fear conditioning (76–79). This enhancement is long-lasting and expressed in the absence of nicotine (79). Conversely, acute nicotine does not enhance the hippocampus-independent association between the auditory CS and the foot-shock US (76,79), even when the difficulty of the task is increased (108). Because nicotine enhances hippocampus-dependent contextual fear conditioning but not hippocampus-independent cued fear conditioning, nicotine may alter hippocampal function or the function of areas that project to the hippocampus during contextual fear conditioning. The neural, cellular, and molecular mechanisms involved in the long-lasting enhancement of contextual conditioning by nicotine are unknown.
Nicotine enhances contextual fear conditioning, and this enhancement is blocked by nAChR antagonists. However, nAChR antagonists administered alone do not disrupt contextual fear conditioning (76–78,109), suggesting that nAChRs mediate the facilitation of contextual fear conditioning but are not essential for this form of learning. Studies with nAChR knockout mice have also suggested that activation of specific nAChR subtypes is not necessary for fear conditioning. Mice lacking either the α7 or the β2 nAChR subunit did not show deficits in conditioned fear (110,111). Neither study examined whether nicotine could enhance fear conditioning in the knockout mice. However, it has since been demonstrated that β2-null mutant mice do not show enhancement of contextual fear conditioning by nicotine. Conversely, α7- and β4-null mutant mice that received nicotine demonstrated enhanced contextual fear conditioning (112). We reported similar results using the α4β2 nAChR antagonist dihydro-β-erythroidine and α7 nAChR antagonist methyllycaconitine; dihydro-β-erythroidine blocked the nicotine enhancement of contextual fear conditioning, but methyllycaconitine did not; neither antagonist disrupted contextual fear conditioning when administered alone (113). Together, these studies indicate that the α4β2 nAChR subtype is involved in the enhancement of contextual conditioning by nicotine.
The neural and molecular mechanisms recruited by nicotine to enhance contextual conditioning are not well known. Long-term memory is believed to be mediated by changes in gene expression that are induced by the activation of intracellular signaling pathways (reviewed in ref. 55). Many of these signaling pathways are activated by calcium. One way in which acute nicotine binding at nAChRs may enhance learning is through interacting with glutamatergic processes to enhance calcium-mediated cell signaling. Nicotine could facilitate glutamate processes thorough presynaptic-mediated glutamate release (114–116). It is also possible that nAChR activation facilitates postsynaptic-mediated glutamate processes involved in learning.
We recently found that co-antagonism of either nAChRs and AMPA glutamate receptors or nAChRs and N-methyl-d-aspartate (NMDA) glutamate receptors with subthreshold doses of antagonists disrupted contextual fear conditioning (117). These findings suggest that nAChRs may mediate processes that are similar or parallel to processes mediated by AMPARs and NMDARs. NMDARs, which are involved in contextual fear conditioning (118–122), require cell depolarization to be activated. It is possible that nicotine enhances contextual fear conditioning by activating nAChRs that contribute to the membrane depolarization necessary for activating NMDAR-mediated processes. For example, nicotine can mediate changes in synaptic current independent of glutamate receptors and can mediate postsynaptic events (93,115,123–131). Additionally, nAChRs have been localized on postsynaptic densities (132,133). These findings suggest that nAChRs can support some fast excitatory synaptic transmission in the absence of glutamate and could play a significant role in NMDAR-dependent synaptic plasticity by contributing to the concurrent membrane depolarization necessary for NMDAR function. Additionally, increased calcium influx mediated by nAChRs (98,99, 134–136) could also facilitate learning by directly activating calcium-mediated cell signaling cascades involved in learning that are also activated by NMDARs.
Multiple studies have shown that nicotine can activate cellular and molecular processes that are involved in the chain of events linking synaptic activity to long-lasting changes associated with gene expression (106,137–141). For example, the MAPK family has been implicated in synaptic plasticity and learning tasks, including contextual conditioning (45,142–147). Nicotine-stimulated changes in calcium influx and in the release of calcium internal stores can activate kinases such as MAPK and CaMKII/IV as well as transcription factors such as CREB (94,103,148,149). In mice, 0.4 mg/kg of acute nicotine increased ERK phosphorylation in area CA2 of the hippocampus (150). Additionally, micro-array studies found nicotine administration was also associated with altered gene expression of several members of the MAPK family (151,152). It remains to be determined whether acute nicotine enhances contextual fear conditioning through enhancement of cell signaling cascades involved in synaptic plasticity.
Chronic Nicotine
As reviewed previously, studies have shown that acute nicotine dose-dependently enhances hippocampus-dependent contextual fear conditioning (76,77,112,153). Because withdrawal effects are commonly the opposite of acute drug action (154), we investigated whether chronic nicotine results in tolerance for the effects of nicotine on contextual fear conditioning and whether withdrawal from chronic nicotine disrupts contextual fear conditioning. We found that an acute dose of nicotine and a chronic dose of nicotine that produced the same plasma nicotine levels did not produce the same behavioral effects; acute nicotine treatment enhanced contextual fear conditioning, whereas chronic nicotine treatment failed to enhance contextual fear conditioning (153). Notably, plasma nicotine levels in mice treated acutely and chronically with nicotine were within the range of plasma nicotine levels (10–50 ng/mL) demonstrated by smokers (155,156). These results suggest that with chronic administration of nicotine, neural adaptation occurs, resulting in tolerance for the effects of nicotine on contextual conditioning; however, the underlying neural changes are unknown.
Although the changes that may contribute to the tolerance observed for effects of nicotine treatment on contextual fear conditioning have not been examined, studies have examined the effects of chronic nicotine on intracellular signaling in multiple brain regions. Chronic nicotine was associated with increased levels of phosphorylated ERK in the prefrontal frontal cortex and decreased ERK levels in the amygdala (157). This study also found that chronic nicotine treatment decreased levels of phosphorylated CREB (pCREB) in the nucleus accumbens and decreased total CREB in the prefrontal cortex. Similarly, another study found that chronic nicotine was associated with increased MAPK activity in the prefrontal cortex (152). It remains to be determined whether chronic nicotine treatment and contextual fear conditioning interact to alter cell-signaling cascades in a manner different than when chronic nicotine is administered without conditioning.
Nicotine Withdrawal
Neural adaptation that occurs during chronic nicotine administration may result in deficits when nicotine is withdrawn. This has been demonstrated for contextual conditioning; mice withdrawn from chronic nicotine treatment demonstrated deficits in contextual fear conditioning compared to their saline-treated counterparts when conditioned 24 h after removal of nicotine (153). It is possible that relapse occurs in smokers after withdrawal from nicotine as an attempt to ameliorate learning-related deficits. For example, an acute challenge dose of nicotine reversed the deficit in contextual conditioning observed in mice withdrawn from chronic nicotine (153).
The neural mechanisms altered during nicotine withdrawal that are responsible for the disruption of contextual fear conditioning are unknown. Changes may occur at both the receptor level and at the level of cell signaling cascades. In support of the former, chronic nicotine exposure was accompanied by an increase in nAChR binding sites (90,158–160) and by nAChR desensitization (90,91,161–163). The increases in nAChR binding sites may be a compensatory mechanism induced by nAChR desensitization (158,162). Evidence from pharmacokinetic studies of nicotine has suggested that periods of nicotine abstinence may result in recovery of function for some of the desensitized nAChRs (for review, see refs. 4, 155, and 156). For example, Gentry and colleagues (164) demonstrated in vitro that desensitized nAChRs recovered function after nicotine removal. Therefore, receptor level changes may account for the behavioral tolerance demonstrated by mice treated chronically with nicotine and for deficits in contextual fear conditioning associated with withdrawal from chronic nicotine.
As reviewed previously, numerous nAChR subtypes exist (for further review, see ref. 165), and alterations in the function and number of any receptor subtype may account for behavioral changes. Research has shown that the acute effects of nicotine on contextual fear conditioning are mediated by α4β2 nAChRs (112,113); however, it is unknown whether the effect of chronic nicotine and withdrawal from chronic nicotine on contextual fear conditioning are also mediated by α4β2 nAChRs. Marks and colleagues (153) reported that chronic nicotine, at a dose that produces comparable plasma nicotine levels as those associated with nicotine withdrawal-related deficits in contextual fear conditioning, results in half-maximal upregulation of α4β2 nAChRs (159). Furthermore, β2-null mutant mice did not show upregulation of nAChRs after chronic nicotine treatment (166). These findings suggest that alterations in α4β2 nAChR function and/or number that occur with chronic nicotine treatment may contribute to the nicotine withdrawal-associated changes in contextual fear conditioning (153). This is an important topic for further study.
In addition to identifying the nAChRs involved in the effects of nicotine withdrawal on contextual fear conditioning, identifying changes in cell signaling cascades that could potentially underlie the effects of nicotine withdrawal on contextual fear conditioning is also important for understanding the effects of nicotine withdrawal on cognition and developing possible therapeutic agents. Studies that have examined the effects of nicotine withdrawal on cell signaling provide potential targets for changes that may cause the nicotine withdrawal-associated deficits in contextual fear conditioning. For example, mecamylamine-precipitated withdrawal from chronic nicotine treatment increased basal and stimulated adenylyl cyclase activity in the amygdala (167). Withdrawal from nicotine may also be associated with changes in activation of gene transcription factors. Twenty-four hours after nicotine withdrawal, increased levels of pCREB were found in the ventral tegmental area and increased levels of total CREB were found in the nucleus accumbens (157). Another series of studies found that 18 h after chronic intermittent nicotine exposure, pCREB was reduced in the cingulated cortex, piriform cortex, parietal cortex, and amygdala (168). Additionally, pCREB levels were also reduced in the shell but not the core area of the nucleus accumbens (57). The difference between this result and that of Brunzell and colleagues (157) may reflect methodological differences in nicotine administration. It remains to be determined whether withdrawal from nicotine differentially alters cell signaling when contextual learning occurs during the withdrawal. We have started experiments to test for possible interactions between nicotine withdrawal and contextual conditioning on cell signaling.
Conclusion
The effects of nicotine on cognition may support the development and maintenance of nicotine addiction through multiple mechanisms. We have demonstrated that acute nicotine enhances contextual learning. Therefore, the initial use of nicotine could facilitate cognitive processes, which may lead to repeated use and to the development of drug–context associations that could precipitate cravings. With repeated use, tolerance for the cognitive enhancing effects of nicotine may also lead to increased use and, therefore, to increased dependence. Finally, withdrawal from chronic nicotine disrupted contextual conditioning in mice. This deficit could be reversed with the administration of acute nicotine (153). In humans, nicotine withdrawal deficits in cognitive function may contribute to relapse in an attempt to ameliorate the deficits. Whereas the α4β2 nAChRs have been identified as the nAChR mediating the acute effects of nicotine on contextual fear conditioning (112,113), the nAChRs involved in the chronic and withdrawal effects of nicotine on contextual fear conditioning are unknown, as are the potential underlying cellular and molecular processes. Identifying the cellular adaptations responsible for behavioral changes in learning that occur with chronic nicotine and withdrawal from chronic nicotine treatment will increase understanding of learning and addiction and may lead to the development of more effective treatments to aid in smoking cessation.
Acknowledgments
Cited work from the author's laboratory was funded in part by a grant from the National Institute on Drug Abuse (DA017949).
References
- 1.Peto R, Lopez AD, Boreham J, Thun M, Heath C, Jr., Doll R. Mortality from smoking worldwide. Br. Med. Bull. 1996;52:12–21. doi: 10.1093/oxfordjournals.bmb.a011519. [DOI] [PubMed] [Google Scholar]
- 2.Corrigall WA. Nicotine self-administration in animals as a dependence model. Nicotine. Tob. Res. 1999;1:11–20. doi: 10.1080/14622299050011121. [DOI] [PubMed] [Google Scholar]
- 3.Rose JE, Corrigall WA. Nicotine self-administration in animals and humans: similarities and differences. Psychopharmacology (Berl.) 1997;130:28–40. doi: 10.1007/s002130050209. [DOI] [PubMed] [Google Scholar]
- 4.Mathieu-Kia AM, Kellogg SH, Butelman ER, Kreek MJ. Nicotine addiction: insights from recent animal studies. Psychopharmacology (Berl.) 2002;162:102–118. doi: 10.1007/s00213-002-1096-0. [DOI] [PubMed] [Google Scholar]
- 5.Malin DH, Lake JR, Newlin-Maultsby P, et al. Rodent model of nicotine abstinence syndrome. Pharmacol. Biochem. Behav. 1992;43:779–784. doi: 10.1016/0091-3057(92)90408-8. [DOI] [PubMed] [Google Scholar]
- 6.Damaj MI, Kao W, Martin BR. Characterization of spontaneous and precipitated nicotine withdrawal in the mouse. J. Pharmacol. Exp. Ther. 2003;307:526–534. doi: 10.1124/jpet.103.054908. [DOI] [PubMed] [Google Scholar]
- 7.Jonkman S, Henry B, Semenova S, Markou A. Mild anxiogenic effects of nicotine withdrawal in mice. Eur. J. Pharmacol. 2005;516:40–45. doi: 10.1016/j.ejphar.2005.04.032. [DOI] [PubMed] [Google Scholar]
- 8.Epping-Jordan MP, Watkins SS, Koob GF, Markou A. Dramatic decreases in brain reward function during nicotine withdrawal. Nature. 1998;393:76–79. doi: 10.1038/30001. [DOI] [PubMed] [Google Scholar]
- 9.Semenova S, Bespalov A, Markou A. Decreased prepulse inhibition during nicotine withdrawal in DBA/2J mice is reversed by nicotine self-administration. Eur. J. Pharmacol. 2003;472:99–110. doi: 10.1016/s0014-2999(03)01904-6. [DOI] [PubMed] [Google Scholar]
- 10.Hughes JR, Gust SW, Skoog K, Keenan RM, Fenwick JW. Symptoms of tobacco withdrawal. A replication and extension. Arch. Gen. Psychiatry. 1991;48:52–59. doi: 10.1001/archpsyc.1991.01810250054007. [DOI] [PubMed] [Google Scholar]
- 11.Kumari V, Gray JA. Smoking withdrawal, nicotine dependence and prepulse inhibition of the acoustic startle reflex. Psychopharmacology (Berl.) 1999;141:11–15. doi: 10.1007/s002130050800. [DOI] [PubMed] [Google Scholar]
- 12.Postma P, Kumari V, Sharma T, Hines M, Gray JA. Startle response during smoking and 24 h after withdrawal predicts successful smoking cessation. Psychopharmacology (Berl.) 2001;156:360–367. doi: 10.1007/s002130100829. [DOI] [PubMed] [Google Scholar]
- 13.Domino EF, Kishimoto T. Tobacco smoking increases gating of irrelevant and enhances attention to relevant tones. Nicotine. Tob. Res. 2002;4:71–78. doi: 10.1080/14622200110098400. [DOI] [PubMed] [Google Scholar]
- 14.Snyder FR, Henningfield JE. Effects of nicotine administration following 12 h of tobacco deprivation: assessment on computerized performance tasks. Psychopharmacology (Berl.) 1989;97:17–22. doi: 10.1007/BF00443406. [DOI] [PubMed] [Google Scholar]
- 15.Kleinman KM, Vaughn RL, Christ TS. Effects of cigarette smoking and smoking deprivation on paired-associate learning of high and low meaningful nonsense syllables. Psychol. Rep. 1973;32:963–966. doi: 10.2466/pr0.1973.32.3.963. [DOI] [PubMed] [Google Scholar]
- 16.Bell SL, Taylor RC, Singleton EG, Henningfield JE, Heishman SJ. Smoking after nicotine deprivation enhances cognitive performance and decreases tobacco craving in drug abusers. Nicotine. Tob. Res. 1999;1:45–52. doi: 10.1080/14622299050011141. [DOI] [PubMed] [Google Scholar]
- 17.Baker TB, Brandon TH, Chassin L. Motivational influences on cigarette smoking. Annu. Rev. Psychol. 2004;55:463–491. doi: 10.1146/annurev.psych.55.090902.142054. [DOI] [PubMed] [Google Scholar]
- 18.Lerman C, Niaura R. Applying genetic approaches to the treatment of nicotine dependence. Oncogene. 2002;21:7412–7420. doi: 10.1038/sj.onc.1205801. [DOI] [PubMed] [Google Scholar]
- 19.Pomerleau CS, Downey KK, Snedecor SM, Mehringer AM, Marks JL, Pomerleau OF. Smoking patterns and abstinence effects in smokers with no ADHD, childhood ADHD, and adult ADHD symptomatology. Add. Behav. 2003;28:1149–1157. doi: 10.1016/s0306-4603(02)00223-x. [DOI] [PubMed] [Google Scholar]
- 20.Adler LE, Olincy A, Waldo M, et al. Schizophrenia, sensory gating, and nicotinic receptors. Schizophr. Bull. 1998;24:189–202. doi: 10.1093/oxfordjournals.schbul.a033320. [DOI] [PubMed] [Google Scholar]
- 21.Kreek MJ, Nielsen DA, Butelman ER, Laforge KS. Genetic influences on impulsivity, risk taking, stress responsivity and vulnerability to drug abuse and addiction. Nat. Neurosci. 2005;8:1450–1457. doi: 10.1038/nn1583. [DOI] [PubMed] [Google Scholar]
- 22.Eichenbaum H. The hippocampus and mechanisms of declarative memory. Behav. Brain Res. 1999;103:123–133. doi: 10.1016/s0166-4328(99)00044-3. [DOI] [PubMed] [Google Scholar]
- 23.Setlow B. The nucleus accumbens and learning and memory. J. Neurosci. Res. 1997;49:515–521. doi: 10.1002/(SICI)1097-4547(19970901)49:5<515::AID-JNR1>3.0.CO;2-E. [DOI] [PubMed] [Google Scholar]
- 24.Blum S, Hebert AE, Dash PK. A role for the prefrontal cortex in recall of recent and remote memories. Neuroreport. 2006;17:341–344. doi: 10.1097/01.wnr.0000201509.53750.bc. [DOI] [PubMed] [Google Scholar]
- 25.Pelletier JG, Pare D. Role of amygdala oscillations in the consolidation of emotional memories. Biol. Psychiatry. 2004;55:559–562. doi: 10.1016/j.biopsych.2003.08.019. [DOI] [PubMed] [Google Scholar]
- 26.Takashima A, Petersson KM, Rutters F, et al. Declarative memory consolidation in humans: a prospective functional magnetic resonance imaging study. Proc. Natl. Acad. Sci. USA. 2006;103:756–761. doi: 10.1073/pnas.0507774103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Eichenbaum H. Declarative memory: insights from cognitive neurobiology. Annu. Rev. Psychol. 1997;48:547–572. doi: 10.1146/annurev.psych.48.1.547. [DOI] [PubMed] [Google Scholar]
- 28.London ED, Ernst M, Grant S, Bonson K, Weinstein A. Orbitofrontal cortex and human drug abuse: functional imaging. Cereb. Cortex. 2000;10:334–342. doi: 10.1093/cercor/10.3.334. [DOI] [PubMed] [Google Scholar]
- 29.Mukhin AG, Gundisch D, Horti AG, et al. 5-Iodo-A-85380, an alpha4beta2 subtype-selective ligand for nicotinic acetylcholine receptors. Mol. Pharmacol. 2000;57:642–649. doi: 10.1124/mol.57.3.642. [DOI] [PubMed] [Google Scholar]
- 30.Bechara A. Decision making, impulse control and loss of willpower to resist drugs: a neurocognitive perspective. Nat. Neurosci. 2005;8:1458–1463. doi: 10.1038/nn1584. [DOI] [PubMed] [Google Scholar]
- 31.See RE. Neural substrates of conditioned-cued relapse to drug-seeking behavior. Pharmacol. Biochem. Behav. 2002;71:517–529. doi: 10.1016/s0091-3057(01)00682-7. [DOI] [PubMed] [Google Scholar]
- 32.Koob GF. Drugs of abuse: anatomy, pharmacology and function of reward pathways. Trends Pharmacol. Sci. 1992;13:177–184. doi: 10.1016/0165-6147(92)90060-j. [DOI] [PubMed] [Google Scholar]
- 33.Di Chiara G, Tanda G, Bassareo V, et al. Drug addiction as a disorder of associative learning—role of nucleus accumbens shell/extended amygdala dopamine. Ann. NY Acad. Sci. 1999;877:461–485. doi: 10.1111/j.1749-6632.1999.tb09283.x. [DOI] [PubMed] [Google Scholar]
- 34.Cornish JL, Kalivas PW. Glutamate transmission in the nucleus accumbens mediates relapse in cocaine addiction. J. Neurosci. 2000;20:RC89. doi: 10.1523/JNEUROSCI.20-15-j0006.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Di Ciano P, Everitt BJ. Dissociable effects of antagonism of NMDA and AMPA/KA receptors in the nucleus accumbens core and shell on cocaine-seeking behavior. Neuropsychopharmacology. 2001;25:341–360. doi: 10.1016/S0893-133X(01)00235-4. [DOI] [PubMed] [Google Scholar]
- 36.Fuchs RA, Evans KA, Ledford CC, et al. The role of the dorsomedial prefrontal cortex, basolateral amygdala, and dorsal hippocampus in contextual reinstatement of cocaine seeking in rats. Neuropsychopharmacology. 2005;30:296–309. doi: 10.1038/sj.npp.1300579. [DOI] [PubMed] [Google Scholar]
- 37.Vorel SR, Liu X, Hayes RJ, Spector JA, Gardner EL. Relapse to cocaine-seeking after hippocampal theta burst stimulation. Science. 2001;292:1175–1178. doi: 10.1126/science.1058043. [DOI] [PubMed] [Google Scholar]
- 38.Robbins TW, Everitt BJ. Limbic-Striatal Memory Systems and Drug Addiction. Neurobiol. Learning Mem. 2002;78:625–636. doi: 10.1006/nlme.2002.4103. [DOI] [PubMed] [Google Scholar]
- 39.Kalivas PW, Volkow ND. The Neural Basis of Addiction: A Pathology of Motivation and Choice. Am. J. Psychiatry. 2005;162:1403–1413. doi: 10.1176/appi.ajp.162.8.1403. [DOI] [PubMed] [Google Scholar]
- 40.Everitt BJ, Robbins TW. Neural systems of reinforcement for drug addiction: from actions to habits to compulsion. Nat. Neurosci. 2005;8:1481–1489. doi: 10.1038/nn1579. [DOI] [PubMed] [Google Scholar]
- 41.Nestler EJ. Common Molecular and Cellular Substrates of Addiction and Memory. Neurobiol. Learn. Mem. 2002;78:637–647. doi: 10.1006/nlme.2002.4084. [DOI] [PubMed] [Google Scholar]
- 42.Pittenger C, Huang YY, Paletzki RF, et al. Reversible inhibition of CREB/ATF transcription factors in region CA1 of the dorsal hippocampus disrupts hippocampus-dependent spatial memory. Neuron. 2002;34:447–462. doi: 10.1016/s0896-6273(02)00684-0. [DOI] [PubMed] [Google Scholar]
- 43.Abel T, Nguyen PV, Barad M, Deuel TA, Kandel ER, Bourtchouladze R. Genetic demonstration of a role for PKA in the late phase of LTP and in hippocampus-based long-term memory. Cell. 1997;88:615–626. doi: 10.1016/s0092-8674(00)81904-2. [DOI] [PubMed] [Google Scholar]
- 44.Selcher JC, Weeber EJ, Varga AW, Sweatt JD, Swank M. Protein kinase signal transduction cascades in mammalian associative conditioning. Neuroscientist. 2002;8:122–131. doi: 10.1177/107385840200800208. [DOI] [PubMed] [Google Scholar]
- 45.Adams JP, Sweatt JD. Molecular psychology: roles for the ERK MAP kinase cascade in memory. Annu. Rev. Pharmacol. Toxicol. 2002;42:135–163. doi: 10.1146/annurev.pharmtox.42.082701.145401. [DOI] [PubMed] [Google Scholar]
- 46.Silva AJ, Kogan JH, Frankland PW, Kida S. CREB and memory. Annu. Rev. Neurosci. 1998;21:127–148. doi: 10.1146/annurev.neuro.21.1.127. [DOI] [PubMed] [Google Scholar]
- 47.Cammarota M, Bevilaqua LR, Viola H, et al. Participation of CaMKII in neuronal plasticity and memory formation. Cell Mol. Neurobiol. 2002;22:259–267. doi: 10.1023/A:1020763716886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Self DW, Genova LM, Hope BT, Barnhart WJ, Spencer JJ, Nestler EJ. Involvement of cAMP-dependent protein kinase in the nucleus accumbens in cocaine self-administration and relapse of cocaine-seeking behavior. J. Neurosci. 1998;18:1848–1859. doi: 10.1523/JNEUROSCI.18-05-01848.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Lynch WJ, Taylor JR. Persistent changes in motivation to self-administer cocaine following modulation of cyclic AMP-dependent protein kinase A (PKA) activity in the nucleus accumbens. Eur. J. Neurosci. 2005;22:1214–1220. doi: 10.1111/j.1460-9568.2005.04305.x. [DOI] [PubMed] [Google Scholar]
- 50.Hou J, Kuromi H, Fukasawa Y, Ueno K, Sakai T, Kidokoro Y. Repetitive exposures to nicotine induce a hyper-responsiveness via the cAMP/PKA/CREB signal pathway in Drosophila. J. Neurobiol. 2004;60:249–261. doi: 10.1002/neu.20021. [DOI] [PubMed] [Google Scholar]
- 51.Pierce RC, Quick EA, Reeder DC, Morgan ZR, Kalivas PW. Calcium-Mediated Second Messengers Modulate the Expression of Behavioral Sensitization to Cocaine. J. Pharmacol. Exp. Ther. 1998;286:1171–1176. [PubMed] [Google Scholar]
- 52.Pierce RC, Pierce-Bancroft AF, Prasad BM. Neurotrophin-3 Contributes to the Initiation of Behavioral Sensitization to Cocaine by Activating the Ras/Mitogen-Activated Protein Kinase Signal Transduction Cascade. J. Neurosci. 1999;19:8685–8695. doi: 10.1523/JNEUROSCI.19-19-08685.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lu L, Hope BT, Dempsey J, Liu SY, Bossert JM, Shaham Y. Central amygdala ERK signaling pathway is critical to incubation of cocaine craving. Nat. Neurosci. 2005;8:212–219. doi: 10.1038/nn1383. [DOI] [PubMed] [Google Scholar]
- 54.Sanna PP, Simpson C, Lutjens R, Koob G. ERK regulation in chronic ethanol exposure and withdrawal. Brain Res. 2002;948:186–191. doi: 10.1016/s0006-8993(02)03191-8. [DOI] [PubMed] [Google Scholar]
- 55.Abel T, Lattal KM. Molecular mechanisms of memory acquisition, consolidation and retrieval. Curr. Opin. Neurobiol. 2001;11:180–187. doi: 10.1016/s0959-4388(00)00194-x. [DOI] [PubMed] [Google Scholar]
- 56.Carlezon J, Duman RS, Nestler EJ. The many faces of CREB. Trends Neurosci. 2005;28:436–445. doi: 10.1016/j.tins.2005.06.005. [DOI] [PubMed] [Google Scholar]
- 57.Pluzarev O, Pandey SC. Modulation of CREB expression and phosphorylation in the rat nucleus accumbens during nicotine exposure and withdrawal. J. Neurosci. Res. 2004;77:884–891. doi: 10.1002/jnr.20216. [DOI] [PubMed] [Google Scholar]
- 58.Carlezon WA, Jr., Thome J, Olson VG, et al. Regulation of Cocaine Reward by CREB. Science. 1998;282:2272–2275. doi: 10.1126/science.282.5397.2272. [DOI] [PubMed] [Google Scholar]
- 59.Widnell KL, Self DW, Lane SB, et al. Regulation of CREB expression: in vivo evidence for a functional role in morphine action in the nucleus accumbens. J. Pharmacol. Exp. Ther. 1996;276:306–315. [PubMed] [Google Scholar]
- 60.Risinger FO, Oakes RA. Nicotine-induced conditioned place preference and conditioned place aversion in mice. Pharmacol. Biochem. Behav. 1995;51:457–461. doi: 10.1016/0091-3057(95)00007-j. [DOI] [PubMed] [Google Scholar]
- 61.Horan B, Smith M, Gardner EL, Lepore M, Ashby CR., Jr (−)-Nicotine produces conditioned place preference in Lewis, but not Fischer 344 rats. Synapse. 1997;26:93–94. doi: 10.1002/(SICI)1098-2396(199705)26:1<93::AID-SYN10>3.0.CO;2-W. [DOI] [PubMed] [Google Scholar]
- 62.Vastola BJ, Douglas LA, Varlinskaya EI, Spear LP. Nicotine-induced conditioned place preference in adolescent and adult rats. Physiol. Behav. 2002;77:107–114. doi: 10.1016/s0031-9384(02)00818-1. [DOI] [PubMed] [Google Scholar]
- 63.Belluzzi JD, Lee AG, Oliff HS, Leslie FM. Age-dependent effects of nicotine on locomotor activity and conditioned place preference in rats. Psychopharmacology (Berl.) 2004;174:389–395. doi: 10.1007/s00213-003-1758-6. [DOI] [PubMed] [Google Scholar]
- 64.Le Foll B, Goldberg SR. Nicotine induces conditioned place preferences over a large range of doses in rats. Psychopharmacology. 2005;178:481–492. doi: 10.1007/s00213-004-2021-5. [DOI] [PubMed] [Google Scholar]
- 65.Grabus S, Martin B, Brown S, Damaj M. Nicotine place preference in the mouse: influences of prior handling, dose and strain and attenuation by nicotinic receptor antagonists. Psychopharmacology. 2006;184:456–463. doi: 10.1007/s00213-006-0305-7. [DOI] [PubMed] [Google Scholar]
- 66.Chaudhri N, Caggiula A, Donny E, Palmatier M, Liu X, Sved A. Complex interactions between nicotine and nonpharmacological stimuli reveal multiple roles for nicotine in reinforcement. Psychopharmacology. 2006;184:353–366. doi: 10.1007/s00213-005-0178-1. [DOI] [PubMed] [Google Scholar]
- 67.Caggiula AR, Donny EC, White AR, et al. Cue dependency of nicotine self-administration and smoking. Pharmacol. Biochem. Behav. 2001;70:515–530. doi: 10.1016/s0091-3057(01)00676-1. [DOI] [PubMed] [Google Scholar]
- 68.Balfour DJ, Wright AE, Benwell ME, Birrell CE. The putative role of extrasynaptic mesolimbic dopamine in the neurobiology of nicotine dependence. Behav. Brain Res. 2000;113:73–83. doi: 10.1016/s0166-4328(00)00202-3. [DOI] [PubMed] [Google Scholar]
- 69.Caggiula AR, Donny EC, White AR, et al. Environmental stimuli promote the acquisition of nicotine self-administration in rats. Psychopharmacology (Berl.) 2002;163:230–237. doi: 10.1007/s00213-002-1156-5. [DOI] [PubMed] [Google Scholar]
- 70.Caggiula AR, Donny EC, Chaudhri N, Perkins KA, Evans-Martin FF, Sved AF. Importance of nonpharmacological factors in nicotine self-administration. Physiol. Behav. 2002;77:683–687. doi: 10.1016/s0031-9384(02)00918-6. [DOI] [PubMed] [Google Scholar]
- 71.Chaudhri N, Caggiula AR, Donny EC, et al. Sex differences in the contribution of nicotine and nonpharmacological stimuli to nicotine self-administration in rats. Psychopharmacology (Berl.) 2005;180:258–266. doi: 10.1007/s00213-005-2152-3. [DOI] [PubMed] [Google Scholar]
- 72.Cohen C, Perrault G, Griebel G, Soubrie P. Nicotine-associated cues maintain nicotine-seeking behavior in rats several weeks after nicotine withdrawal: reversal by the cannabinoid (CB1) receptor antagonist, rimonabant (SR141716) Neuropsychopharmacology. 2005;30:145–155. doi: 10.1038/sj.npp.1300541. [DOI] [PubMed] [Google Scholar]
- 73.White NM. Addictive drugs as reinforcers: multiple partial actions on memory systems. Addiction. 1996;91:921–949. [PubMed] [Google Scholar]
- 74.Logue SF, Paylor R, Wehner JM. Hippocampal lesions cause learning deficits in inbred mice in the Morris water maze and conditioned-fear task. Behav. Neurosci. 1997;111:104–113. doi: 10.1037//0735-7044.111.1.104. [DOI] [PubMed] [Google Scholar]
- 75.Phillips RG, Ledoux JE. Differential contribution of amygdala and hippocampus to cued and contextual fear conditioning. Behav. Neurosci. 1992;106:274–285. doi: 10.1037//0735-7044.106.2.274. [DOI] [PubMed] [Google Scholar]
- 76.Gould TJ, Wehner JM. Nicotine enhancement of contextual fear conditioning. Behav. Brain Res. 1999;102:31–39. doi: 10.1016/s0166-4328(98)00157-0. [DOI] [PubMed] [Google Scholar]
- 77.Gould TJ. Nicotine produces a within subject enhancement of contextual fear conditioning in C57BL/6 mice independent of sex. Int. Physiol. Behav. Science. 2003;38:124–132. doi: 10.1007/BF02688830. [DOI] [PubMed] [Google Scholar]
- 78.Gould TJ, Lommock JA. Nicotine enhances contextual fear conditioning and ameliorates ethanol-induced deficits in contextual fear conditioning. Behav. Neurosci. 2003;117:1276–1282. doi: 10.1037/0735-7044.117.6.1276. [DOI] [PubMed] [Google Scholar]
- 79.Gould TJ, Higgins JS. Nicotine enhances contextual fear conditioning in C57BL/6J mice at 1 and 7 days post-training. Neurobiol. Learn. Mem. 2003;80:147–157. doi: 10.1016/s1074-7427(03)00057-1. [DOI] [PubMed] [Google Scholar]
- 80.Rossebo OE, Gould TJ. Nicotine enhancement of cued trace fear conditioning but not cued delay fear conditioning in C57BL/6J mice. Soc. Neurosci. Abs. 2003;89:1. [Google Scholar]
- 81.Levin ED, Rose JE. Nicotinic and muscarinic interactions and choice accuracy in the radial-arm maze. Brain Res. Bull. 1991;27:125–128. doi: 10.1016/0361-9230(91)90293-s. [DOI] [PubMed] [Google Scholar]
- 82.Rush R, Kuryatov A, Nelson ME, Lindstrom J. First and second transmembrane segments of alpha3, alpha4, beta2, and beta4 nicotinic acetylcholine receptor subunits influence the efficacy and potency of nicotine. Mol. Pharmacol. 2002;61:1416–1422. doi: 10.1124/mol.61.6.1416. [DOI] [PubMed] [Google Scholar]
- 83.Lena C, Changeux JP. Allosteric nicotinic receptors, human pathologies. J. Physiol Paris. 1998;92:63–74. doi: 10.1016/S0928-4257(98)80140-X. [DOI] [PubMed] [Google Scholar]
- 84.le Novere N, Grutter T, Changeux JP. Models of the extracellular domain of the nicotinic receptors and of agonist- and Ca2+-binding sites. Proc. Natl. Acad. Sci. USA. 2002;99:3210–3215. doi: 10.1073/pnas.042699699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Cordero-Erausquin M, Marubio LM, Klink R, Changeux JP. Nicotinic receptor function: new perspectives from knockout mice. Trends Pharmacol. Sci. 2000;21:211–217. doi: 10.1016/s0165-6147(00)01489-9. [DOI] [PubMed] [Google Scholar]
- 86.Broide RS, Leslie FM. The alpha7 nicotinic acetylcholine receptor in neuronal plasticity. Mol. Neurobiol. 1999;20:1–16. doi: 10.1007/BF02741361. [DOI] [PubMed] [Google Scholar]
- 87.Wonnacott S. Presynaptic nicotinic ACh receptors. Trends. Neurosci. 1997;20:92–98. doi: 10.1016/s0166-2236(96)10073-4. [DOI] [PubMed] [Google Scholar]
- 88.Perry DC, Xiao Y, Nguyen HN, Musachio JL, Davila-Garcia MI, Kellar KJ. Measuring nicotinic receptors with characteristics of alpha4beta2, alpha3beta2 and alpha3beta4 subtypes in rat tissues by autoradiography. J. Neurochem. 2002;82:468–481. doi: 10.1046/j.1471-4159.2002.00951.x. [DOI] [PubMed] [Google Scholar]
- 89.Papke RL, Sanberg PR, Shytle RD. Analysis of mecamylamine stereoisomers on human nicotinic receptor subtypes. J. Pharmacol. Exp. Ther. 2001;297:646–656. [PubMed] [Google Scholar]
- 90.Fenster CP, Rains MF, Noerager B, Quick MW, Lester RA. Influence of subunit composition on desensitization of neuronal acetylcholine receptors at low concentrations of nicotine. J. Neurosci. 1997;17:5747–5759. doi: 10.1523/JNEUROSCI.17-15-05747.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wooltorton JR, Pidoplichko VI, Broide RS, Dani JA. Differential desensitization and distribution of nicotinic acetylcholine receptor subtypes in midbrain dopamine areas. J. Neurosci. 2003;23:3176–3185. doi: 10.1523/JNEUROSCI.23-08-03176.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Orr-Urtreger A, Goldner FM, Saeki M, et al. Mice deficient in the alpha7 neuronal nicotinic acetylcholine receptor lack alpha-bungarotoxin binding sites and hippocampal fast nicotinic currents. J. Neurosci. 1997;17:9165–9171. doi: 10.1523/JNEUROSCI.17-23-09165.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Zhang ZW, Coggan JS, Berg DK. Synaptic currents generated by neuronal acetylcholine receptors sensitive to alpha-bungarotoxin. Neuron. 1996;17:1231–1240. doi: 10.1016/s0896-6273(00)80253-6. [DOI] [PubMed] [Google Scholar]
- 94.Berg DK, Conroy WG. Nicotinic alpha 7 receptors: synaptic options and downstream signaling in neurons. J. Neurobiol. 2002;53:512–523. doi: 10.1002/neu.10116. [DOI] [PubMed] [Google Scholar]
- 95.Sorenson EM, Shiroyama T, Kitai ST. Postsynaptic nicotinic receptors on dopaminergic neurons in the substantia nigra pars compacta of the rat. Neuroscience. 1998;87:659–673. doi: 10.1016/s0306-4522(98)00064-5. [DOI] [PubMed] [Google Scholar]
- 96.Porter JT, Cauli B, Tsuzuki K, Lambolez B, Rossier J, Audinat E. Selective excitation of subtypes of neocortical interneurons by nicotinic receptors. J. Neurosci. 1999;19:5228–5235. doi: 10.1523/JNEUROSCI.19-13-05228.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Nomikos GG, Schilstrom B, Hildebrand BE, Panagis G, Grenhoff J, Svensson TH. Role of alpha7 nicotinic receptors in nicotine dependence and implications for psychiatric illness. Behav. Brain Res. 2000;113:97–103. doi: 10.1016/s0166-4328(00)00204-7. [DOI] [PubMed] [Google Scholar]
- 98.Eilers H, Schaeffer E, Bickler PE, Forsayeth JR. Functional deactivation of the major neuronal nicotinic receptor caused by nicotine and a protein kinase C-dependent mechanism. Mol. Pharmacol. 1997;52:1105–1112. [PubMed] [Google Scholar]
- 99.Dajas-Bailador FA, Mogg AJ, Wonnacott S. Intracellular Ca2+ signals evoked by stimulation of nicotinic acetylcholine receptors in SH-SY5Y cells: contribution of voltage operated Ca2+ channels and Ca2+ stores. J. Neurochem. 2002;81:606–614. doi: 10.1046/j.1471-4159.2002.00846.x. [DOI] [PubMed] [Google Scholar]
- 100.Barrantes GE, Murphy CT, Westwick J, Wonnacott S. Nicotine increases intracellular calcium in rat hippocampal neurons via voltage-gated calcium channels. Neurosci. Lett. 1995;196:101–104. doi: 10.1016/0304-3940(95)11859-u. [DOI] [PubMed] [Google Scholar]
- 101.Barrantes GE, Westwick J, Wonnacott S. Nicotinic acetylcholine receptors in primary cultures of hippocampal neurons: pharmacology and Ca++ permeability. Biochem. Soc. Trans. 1994;22 doi: 10.1042/bst022294s. 294S. [DOI] [PubMed] [Google Scholar]
- 102.Sabban EL, Gueorguiev VD. Effects of short- and long-term nicotine treatment on intracellular calcium and tyrosine hydroxylase gene expression. Ann. NY Acad. Sci. 2002;971:39–44. doi: 10.1111/j.1749-6632.2002.tb04430.x. [DOI] [PubMed] [Google Scholar]
- 103.Nakayama H, Numakawa T, Ikeuchi T, Hatanaka H. Nicotine-induced phosphorylation of extracellular signal-regulated protein kinase and CREB in PC12h cells. J. Neurochem. 2001;79:489–498. doi: 10.1046/j.1471-4159.2001.00602.x. [DOI] [PubMed] [Google Scholar]
- 104.Grottick AJ, Higgins GA. Effect of subtype selective nicotinic compounds on attention as assessed by the five-choice serial reaction time task. Behav. Brain Res. 2000;117:197–208. doi: 10.1016/s0166-4328(00)00305-3. [DOI] [PubMed] [Google Scholar]
- 105.Grillner P, Svensson TH. Nicotine-induced excitation of midbrain dopamine neurons in vitro involves ionotropic glutamate receptor activation. Synapse. 2000;38:1–9. doi: 10.1002/1098-2396(200010)38:1<1::AID-SYN1>3.0.CO;2-A. [DOI] [PubMed] [Google Scholar]
- 106.Fujii S, Ji Z, Sumikawa K. Inactivation of alpha7 ACh receptors and activation of non-alpha7 ACh receptors both contribute to long term potentiation induction in the hippocampal CA1 region. Neurosci. Lett. 2000;286:134–138. doi: 10.1016/s0304-3940(00)01076-4. [DOI] [PubMed] [Google Scholar]
- 107.Maren S. Neurobiology of Pavlovian fear conditioning. Annu. Rev. Neurosci. 2001;24:897–931. doi: 10.1146/annurev.neuro.24.1.897. [DOI] [PubMed] [Google Scholar]
- 108.Gould TJ, Feiro O, Moore D. Nicotine enhancement of trace cued fear conditioning but not delay cued fear conditioning in C57BL/6J mice. Behav. Brain Res. 2004;155:167–173. doi: 10.1016/j.bbr.2004.04.009. [DOI] [PubMed] [Google Scholar]
- 109.Feiro O, Gould TJ. The interactive effects of nicotinic and muscarinic cholinergic receptor inhibition on fear conditioning in young and aged C57BL/6 mice. Pharmacol. Biochem. Behav. 2005;80:251–262. doi: 10.1016/j.pbb.2004.11.005. [DOI] [PubMed] [Google Scholar]
- 110.Caldarone BJ, Duman CH, Picciotto MR. Fear conditioning and latent inhibition in mice lacking the high affinity subclass of nicotinic acetylcholine receptors in the brain. Neuropharmacology. 2000;39:2779–2784. doi: 10.1016/s0028-3908(00)00137-4. [DOI] [PubMed] [Google Scholar]
- 111.Paylor R, Nguyen M, Crawley JN, Patrick J, Beaudet A, Orr-Urtreger A. Alpha 7 nicotinic receptor subunits are not necessary for hippocampal-dependent learning or sensorimotor gating: a behavioral characterization of Acra7-deficient mice. Learn. Mem. 1998;5:302–316. [PMC free article] [PubMed] [Google Scholar]
- 112.Wehner JM, Keller JJ, Keller AB, et al. Role of neuronal nicotinic receptors in the effects of nicotine and ethanol on contextual fear conditioning. Neuroscience. 2004;129:11–24. doi: 10.1016/j.neuroscience.2004.07.016. [DOI] [PubMed] [Google Scholar]
- 113.Davis JA, Gould TJ. The effects of DHBE and MLA on nicotine-induced enhancement of contextual fear conditioning in C57BL/6 mice. Psychopharmacology. 2006;184:345–352. doi: 10.1007/s00213-005-0047-y. [DOI] [PubMed] [Google Scholar]
- 114.Wu M, Hajszan T, Leranth C, Alreja M. Nicotine recruits a local glutamatergic circuit to excite septohippocampal GABAergic neurons. Eur. J. Neurosci. 2003;18:1155–1168. doi: 10.1046/j.1460-9568.2003.02847.x. [DOI] [PubMed] [Google Scholar]
- 115.Alkondon M, Pereira EFR, Albuquerque EX. NMDA and AMPA Receptors Contribute to the Nicotinic Cholinergic Excitation of CA1 Interneurons in the Rat Hippocampus. J. Neurophysiol. 2003;90:1613–1625. doi: 10.1152/jn.00214.2003. [DOI] [PubMed] [Google Scholar]
- 116.Radcliffe KA, Fisher JL, Gray R, Dani JA. Nicotinic modulation of glutamate and GABA synaptic transmission of hippocampal neurons. Ann. NY Acad. Sci. 1999;868:591–610. doi: 10.1111/j.1749-6632.1999.tb11332.x. [DOI] [PubMed] [Google Scholar]
- 117.Gould TJ, Lewis MC. Coantagonism of glutamate receptors and nicotinic acetylcholinergic receptors disrupts fear conditioning and latent inhibition of fear conditioning. Learn. Mem. 2005;12:389–398. doi: 10.1101/lm.89105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Kim JJ, DeCola JP, Landeira-Fernandez J, Fanselow MS. N-methyl-D-aspartate receptor antagonist APV blocks acquisition but not expression of fear conditioning. Behav. Neurosci. 1991;105:126–133. doi: 10.1037//0735-7044.105.1.126. [DOI] [PubMed] [Google Scholar]
- 119.Fanselow MS, Kim JJ, Yipp J, De Oca B. Differential effects of the N-methyl-D-aspartate antagonist DL-2-amino-5-phosphonovalerate on acquisition of fear of auditory and contextual cues. Behav. Neurosci. 1994;108:235–240. doi: 10.1037//0735-7044.108.2.235. [DOI] [PubMed] [Google Scholar]
- 120.Fanselow MS, Kim JJ. Acquisition of contextual Pavlovian fear conditioning is blocked by application of an NMDA receptor antagonist D,L-2-amino-5-phosphonovaleric acid to the basolateral amygdala. Behav. Neurosci. 1994;108:210–212. doi: 10.1037//0735-7044.108.1.210. [DOI] [PubMed] [Google Scholar]
- 121.Gould TJ, McCarthy MM, Keith RA. MK-801 disrupts acquisition of contextual fear conditioning but enhances memory consolidation of cued fear conditioning. Behav. Pharmacol. 2002;13:287–294. doi: 10.1097/00008877-200207000-00005. [DOI] [PubMed] [Google Scholar]
- 122.Stiedl O, Birkenfeld K, Palve M, Spiess J. Impairment of conditioned contextual fear of C57BL/6J mice by intracerebral injections of the NMDA receptor antagonist APV. Behav. Brain Res. 2000;116:157–168. doi: 10.1016/s0166-4328(00)00269-2. [DOI] [PubMed] [Google Scholar]
- 123.Alkondon M, Pereira EF, Albuquerque EX. alpha-bungarotoxin-and methyllycaconitine-sensitive nicotinic receptors mediate fast synaptic transmission in interneurons of rat hippocampal slices. Brain Res. 1998;810:257–263. doi: 10.1016/s0006-8993(98)00880-4. [DOI] [PubMed] [Google Scholar]
- 124.Frazier CJ, Buhler AV, Weiner JL, Dunwiddie TV. Synaptic Potentials Mediated via alpha -Bungarotoxin-Sensitive Nicotinic Acetylcholine Receptors in Rat Hippocampal Interneurons. J. Neurosci. 1998;18:8228–8235. doi: 10.1523/JNEUROSCI.18-20-08228.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Hefft S, Hulo S, Bertrand D, Muller D. Synaptic transmission at nicotinic acetylcholine receptors in rat hippocampal organotypic cultures and slices. J. Physiol. (Lond.) 1999;515:769–776. doi: 10.1111/j.1469-7793.1999.769ab.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Matsubayashi H, Inoue A, Amano T, et al. Involvement of (alpha)7- and (alpha)4(beta)2-type postsynaptic nicotinic acetylcholine receptors in nicotine-induced excitation of dopaminergic neurons in the substantia nigra: a patch clamp and single-cell PCR study using acutely dissociated nigral neurons. Mol. Brain Res. 2004;129:1–7. doi: 10.1016/j.molbrainres.2004.06.040. [DOI] [PubMed] [Google Scholar]
- 127.Matsubayashi H, Amano T, Seki T, Sasa M, Sakai N. Postsynaptic (alpha)4(beta)2 and (alpha)7 type nicotinic acetylcholine receptors contribute to the local and endogenous acetylcholine-mediated synaptic transmissions in nigral dopaminergic neurons. Brain Res. 2004;1005:1–8. doi: 10.1016/j.brainres.2004.01.040. [DOI] [PubMed] [Google Scholar]
- 128.Chen Y, Sharples TJW, Phillips KG, et al. The nicotinic (alpha)4(beta)2 receptor selective agonist, TC-2559, increases dopamine neuronal activity in the ventral tegmental area of rat midbrain slices. Neuropharmacology. 2003;45:334–344. doi: 10.1016/s0028-3908(03)00189-8. [DOI] [PubMed] [Google Scholar]
- 129.Ji D, Lape R, Dani JA. Timing and Location of Nicotinic Activity Enhances or Depresses Hippocampal Synaptic Plasticity. Neuron. 2001;31:131–141. doi: 10.1016/s0896-6273(01)00332-4. [DOI] [PubMed] [Google Scholar]
- 130.Roerig B, Nelson DA, Katz LC. Fast Synaptic Signaling by Nicotinic Acetylcholine and Serotonin 5-HT3 Receptors in Developing Visual Cortex. J. Neurosci. 1997;17:8353–8362. doi: 10.1523/JNEUROSCI.17-21-08353.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Chu ZG, Zhou FM, Hablitz JJ. Nicotinic acetylcholine receptor-mediated synaptic potentials in rat neocortex. Brain Res. 2000;887:399–405. doi: 10.1016/s0006-8993(00)03076-6. [DOI] [PubMed] [Google Scholar]
- 132.Fabian-Fine R, Skehel P, Errington ML, et al. Ultrastructural Distribution of the α7 Nicotinic Acetylcholine Receptor Subunit in Rat Hippocampus. J. Neurosci. 2001;21:7993–8003. doi: 10.1523/JNEUROSCI.21-20-07993.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Levy RB, Aoki C. Alpha7 nicotinic acetylcholine receptors occur at postsynaptic densities of AMPA receptor-positive and-negative excitatory synapses in rat sensory cortex. J. Neurosci. 2002;22:5001–5015. doi: 10.1523/JNEUROSCI.22-12-05001.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Broide RS, Leslie FM. The alpha7 nicotinic acetylcholine receptor in neuronal plasticity. Mol. Neurobiol. 1999;20:1–16. doi: 10.1007/BF02741361. [DOI] [PubMed] [Google Scholar]
- 135.Chavez-Noriega LE, Gillespie A, Stauderman KA, et al. Characterization of the recombinant human neuronal nicotinic acetylcholine receptors (alpha)3(beta)2 and (alpha)4(beta)2 stably expressed in HEK293 cells*1. Neuropharmacology. 2000;39:2543–2560. doi: 10.1016/s0028-3908(00)00134-9. [DOI] [PubMed] [Google Scholar]
- 136.Karadsheh MS, Shah MS, Tang X, Macdonald RL, Stitzel JA. Functional characterization of mouse alpha4beta2 nicotinic acetylcholine receptors stably expressed in HEK293T cells. J. Neurochem. 2004;91:1138–1150. doi: 10.1111/j.1471-4159.2004.02801.x. [DOI] [PubMed] [Google Scholar]
- 137.Fujii S, Sumikawa K. Acute and chronic nicotine exposure reverse age-related declines in the induction of long-term potentiation in the rat hippocampus. Brain Res. 2001;894:347–353. doi: 10.1016/s0006-8993(01)02057-1. [DOI] [PubMed] [Google Scholar]
- 138.Fujii S, Sumikawa K. Nicotine accelerates reversal of long-term potentiation and enhances long-term depression in the rat hippocampal CA1 region. Brain Res. 2001;894:340–346. doi: 10.1016/s0006-8993(01)02058-3. [DOI] [PubMed] [Google Scholar]
- 139.Radcliffe KA, Dani JA. Nicotinic stimulation produces multiple forms of increased glutamatergic synaptic transmission. J. Neurosci. 1998;18:7075–7083. doi: 10.1523/JNEUROSCI.18-18-07075.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Fujii S, Jia Y, Yang A, Sumikawa K. Nicotine reverses GABAergic inhibition of long-term potentiation induction in the hippocampal CA1 region. Brain Res. 2000;863:259–265. doi: 10.1016/s0006-8993(00)02119-3. [DOI] [PubMed] [Google Scholar]
- 141.Fisher JL, Dani JA. Nicotinic receptors on hippocampal cultures can increase synaptic glutamate currents while decreasing the NMDA-receptor component. Neuropharmacology. 2000;39:2756–2769. doi: 10.1016/s0028-3908(00)00102-7. [DOI] [PubMed] [Google Scholar]
- 142.Alonso M, Bevilaqua LR, Izquierdo I, Medina JH, Cammarota M. Memory formation requires p38MAPK activity in the rat hippocampus. Neuroreport. 2003;14:1989–1992. doi: 10.1097/00001756-200310270-00022. [DOI] [PubMed] [Google Scholar]
- 143.Atkins CM, Selcher JC, Petraitis JJ, Trzaskos JM, Sweatt JD. The MAPK cascade is required for mammalian associative learning. Nat. Neurosci. 1998;1:602–609. doi: 10.1038/2836. [DOI] [PubMed] [Google Scholar]
- 144.Waltereit R, Weller M. Signaling from cAMP/PKA to MAPK and synaptic plasticity. Mol. Neurobiol. 2003;27:99–106. doi: 10.1385/MN:27:1:99. [DOI] [PubMed] [Google Scholar]
- 145.Schafe GE, Atkins CM, Swank MW, Bauer EP, Sweatt JD, Ledoux JE. Activation of ERK/MAP kinase in the amygdala is required for memory consolidation of pavlovian fear conditioning. J. Neurosci. 2000;20:8177–8187. doi: 10.1523/JNEUROSCI.20-21-08177.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Selcher JC, Atkins CM, Trzaskos JM, Paylor R, Sweatt JD. A necessity for MAP kinase activation in mammalian spatial learning. Learn. Mem. 1999;6:478–490. doi: 10.1101/lm.6.5.478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Shalin SC, Zirrgiebel U, Honsa KJ, et al. Neuronal MEK is important for normal fear conditioning in mice. J. Neurosci. Res. 2004;75:760–770. doi: 10.1002/jnr.20052. [DOI] [PubMed] [Google Scholar]
- 148.Chang KT, Berg DK. Voltage-gated channels block nicotinic regulation of CREB phosphorylation and gene expression in neurons. Neuron. 2001;32:855–865. doi: 10.1016/s0896-6273(01)00516-5. [DOI] [PubMed] [Google Scholar]
- 149.Dajas-Bailador FA, Soliakov L, Wonnacott S. Nicotine activates the extracellular signal-regulated kinase 1/2 via the alpha7 nicotinic acetylcholine receptor and protein kinase A, in SH-SY5Y cells and hippocampal neurones. J. Neurochem. 2002;80:520–530. doi: 10.1046/j.0022-3042.2001.00725.x. [DOI] [PubMed] [Google Scholar]
- 150.Valjent E, Pages C, Herve D, Girault JA, Caboche J. Addictive and non-addictive drugs induce distinct and specific patterns of ERK activation in mouse brain. Eur. J. Neurosci. 2004;19:1826–1836. doi: 10.1111/j.1460-9568.2004.03278.x. [DOI] [PubMed] [Google Scholar]
- 151.Zhang SH, Day IM, Ye SH. Microarray analysis of nicotine-induced changes in gene expression in endothelial cells. Physiol. Genom. 2001;5:187–192. doi: 10.1152/physiolgenomics.2001.5.4.187. [DOI] [PubMed] [Google Scholar]
- 152.Konu O, Kane JK, Barrett T, et al. Region-specific transcriptional response to chronic nicotine in rat brain. Brain Res. 2001;909:194–203. doi: 10.1016/s0006-8993(01)02685-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Davis JA, James JR, Siegel SJ, Gould TJ. Withdrawal from chronic nicotine administration impairs contextual fear conditioning in C57BL/6 mice. J. Neurosci. 2005;25:8708–8713. doi: 10.1523/JNEUROSCI.2853-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Koob GF, Bloom FE. Cellular and molecular mechanisms of drug dependence. Science. 1988;242:715–723. doi: 10.1126/science.2903550. [DOI] [PubMed] [Google Scholar]
- 155.Benowitz NL, Porchet H, Jacob P., III Nicotine dependence and tolerance in man: pharmacokinetic and pharmacodynamic investigations. Prog. Brain Res. 1989;79:279–287. doi: 10.1016/s0079-6123(08)62487-5. [DOI] [PubMed] [Google Scholar]
- 156.Henningfield JE, Keenan RM. Nicotine delivery kinetics and abuse liability. J. Consult. Clin. Psychol. 1993;61:743–750. doi: 10.1037//0022-006x.61.5.743. [DOI] [PubMed] [Google Scholar]
- 157.Brunzell DH, Russell DS, Picciotto MR. In vivo nicotine treatment regulates mesocorticolimbic CREB and ERK signaling in C57Bl/6J mice. J. Neurochem. 2003;84:1431–1441. doi: 10.1046/j.1471-4159.2003.01640.x. [DOI] [PubMed] [Google Scholar]
- 158.Marks MJ, Burch JB, Collins AC. Genetics of nicotine response in four inbred strains of mice. J. Pharmacol. Exp. Ther. 1983;226:291–302. [PubMed] [Google Scholar]
- 159.Marks MJ, Rowell PP, Cao JZ, Grady SR, McCallum SE, Collins AC. Subsets of acetylcholine-stimulated 86Rb+ efflux and (125I)-epibatidine binding sites in C57BL/6 mouse brain are differentially affected by chronic nicotine treatment. Neuropharmacology. 2004;46:1141–1157. doi: 10.1016/j.neuropharm.2004.02.009. [DOI] [PubMed] [Google Scholar]
- 160.Peng X, Gerzanich V, Anand R, Wang F, Lindstrom J. Chronic nicotine treatment up-regulates alpha3 and alpha7 acetylcholine receptor subtypes expressed by the human neuroblastoma cell line SH-SY5Y. Mol. Pharmacol. 1997;51:776–784. doi: 10.1124/mol.51.5.776. [DOI] [PubMed] [Google Scholar]
- 161.Marks MJ, Grady SR, Collins AC. Downregulation of nicotinic receptor function after chronic nicotine infusion. J. Pharmacol. Exp. Ther. 1993;266:1268–1276. [PubMed] [Google Scholar]
- 162.Schwartz RD, Kellar KJ. In vivo regulation of (3H)acetylcholine recognition sites in brain by nicotinic cholinergic drugs. J. Neurochem. 1985;45:427–433. doi: 10.1111/j.1471-4159.1985.tb04005.x. [DOI] [PubMed] [Google Scholar]
- 163.Olale F, Gerzanich V, Kuryatov A, Wang F, Lindstrom J. Chronic nicotine exposure differentially affects the function of human alpha3, alpha4, and alpha7 neuronal nicotinic receptor subtypes. J. Pharmacol. Exp. Ther. 1997;283:675–683. [PubMed] [Google Scholar]
- 164.Gentry CL, Wilkins LH, Jr., Lukas RJ. Effects of Prolonged Nicotinic Ligand Exposure on Function of Heterologously Expressed, Human alpha 4beta 2- and alpha 4beta 4-Nicotinic Acetylcholine Receptors. J. Pharmacol. Exp. Ther. 2003;304:206–216. doi: 10.1124/jpet.102.041756. [DOI] [PubMed] [Google Scholar]
- 165.Picciotto MR, Caldarone BJ, King SL, Zachariou V. Nicotinic receptors in the brain. Links between molecular biology and behavior. Neuropsychopharmacology. 2000;22:451–465. doi: 10.1016/S0893-133X(99)00146-3. [DOI] [PubMed] [Google Scholar]
- 166.McCallum S, Collins A, Paylor R, Marks M. Deletion of the beta 2 nicotinic acetylcholine receptor subunit alters development of tolerance to nicotine and eliminates receptor upregulation. Psychopharmacology. 2006;184:314–327. doi: 10.1007/s00213-005-0076-6. [DOI] [PubMed] [Google Scholar]
- 167.Tzavara ET, Monory K, Hanoune J, Nomikos GG. Nicotine withdrawal syndrome: behavioural distress and selective up-regulation of the cyclic AMP pathway in the amygdala. Eur. J. Neurosci. 2002;16:149–153. doi: 10.1046/j.1460-9568.2002.02061.x. [DOI] [PubMed] [Google Scholar]
- 168.Pandey SC, Roy A, Xu T, Mittal N. Effects of protracted nicotine exposure and withdrawal on the expression and phosphorylation of the CREB gene transcription factor in rat brain. J. Neurochem. 2001;77:943–952. doi: 10.1046/j.1471-4159.2001.00309.x. [DOI] [PubMed] [Google Scholar]